EP3712151B1 - (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine en tant qu'inhibiteur de shp2 pour le traitement du cancer - Google Patents

(s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine en tant qu'inhibiteur de shp2 pour le traitement du cancer Download PDF

Info

Publication number
EP3712151B1
EP3712151B1 EP19881313.1A EP19881313A EP3712151B1 EP 3712151 B1 EP3712151 B1 EP 3712151B1 EP 19881313 A EP19881313 A EP 19881313A EP 3712151 B1 EP3712151 B1 EP 3712151B1
Authority
EP
European Patent Office
Prior art keywords
compound
mixture
reaction
dihydrospiro
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP19881313.1A
Other languages
German (de)
English (en)
Other versions
EP3712151A1 (fr
EP3712151A4 (fr
Inventor
Qiangang ZHENG
Ming Xu
Qinglong Zeng
Jing Li
Hao ZHUGE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Etern Biopharma Shanghai Co Ltd
Original Assignee
Etern Biopharma Shanghai Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=70516741&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP3712151(B1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Etern Biopharma Shanghai Co Ltd filed Critical Etern Biopharma Shanghai Co Ltd
Priority to EP22159530.9A priority Critical patent/EP4074713A1/fr
Publication of EP3712151A1 publication Critical patent/EP3712151A1/fr
Publication of EP3712151A4 publication Critical patent/EP3712151A4/fr
Application granted granted Critical
Publication of EP3712151B1 publication Critical patent/EP3712151B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • This disclosure relates to spiro aromatic ring compounds.
  • the disclosure relates to spiro aromatic ring compounds which can be used as an SHP2 inhibitor, the pharmaceutically acceptable salts thereof, or enantiomers, diastereoisomers, tautomers, solvates or isotope-substituted derivatives thereof.
  • the disclosure also relates to methods for the preparation of the compounds, pharmaceutical compositions comprising the compounds and use of the compounds in the preparation of medicaments for the prevention or treatment of diseases or conditions related to abnormal SHP2 activity.
  • Protein tyrosine phosphatase SHP2 plays an important role in cell signaling and is a target for the treatment of major diseases such as diabetes, autoimmune diseases and cancers.
  • SHP2 is mutated or highly expressed in various diseases, such as Noonan Syndrome, Leopard Syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of head and neck, gastric cancer, anaplastic large cell lymphoma, and glioblastoma, etc.
  • SHP2 is involved in multiple tumor cell signaling pathways, such as MAPK, JAK/STAT, and PI3K/Akt, etc.
  • SHP2 is also responsible for the signal transduction of PD1-PDL1 immunosuppressive pathway. Therefore, inhibition of SHP2 activity can reverse immunosuppression in tumor microenvironment.
  • SHP2 consists of two N-terminal Src Homolgy-2 domains (N-SH2 and C-SH2) and a protein tyrosine phosphatase catalytic domain (PTP).
  • N-SH2 combines with PTP to form a ring structure, which hinders the binding of PTP to substrate, thus inhibiting the enzyme catalytic activity; when the tyrosine of an upstream receptor protein is phosphorylated and binds to N-SH2, PTP catalytic domain is released to exhibit phosphatase activity.
  • SHP2 inhibitors mainly focus on allosteric inhibitors in the non-catalytic region, such as the compounds disclosed in WO2015107493A1 , WO2016203404A1 , WO2016203406A1 , WO2017216706A1 , WO2017211303A1 , CN201710062495 , WO2018136265A1 , WO2018057884 , etc.
  • Heterocyclic derivatives useful as SHP2 inhibitors are disclosed e.g., in WO 2018172984 .
  • Compounds and compositions for inhibiting the activity of SHP2 are disclosed e.g., in CN 105 916 845 and CN 105 899 491 .
  • the present invention is directed to a compound having a structure: or a pharmaceutically acceptable salt, an enantiomer, diastereoisomer, tautomer, solvate, or isotope-substituted derivative thereof. Furthermore, the present invention is directed to a pharmaceutical composition comprising:
  • One of X 1 and X 2 may be CH 2 , and the other may be a bond.
  • X 3 may be S.
  • X 4 may be selected from N or CH.
  • R 1 , R 2 , R 3 , R 4 and R 7 may be independently selected from H, -OH, -F, -Cl, -Br, -NH 2 , -NHC 1-3 alkyl, methyl, ethyl, propyl, isopropyl, butyl, methoxy, ethoxy, propoxy or isopropoxy; C 1-3 alkyl substituted by halogen, -NH 2 , -OH, C 1-3 alkyl or C 1-3 alkoxyl; or C 1-3 alkoxyl substituted by halogen, -NH 2 , -OH, C 1-3 alkyl or C 1-3 alkoxyl.
  • R 5 and R 6 may be independently selected from H, -OH, -F, -Cl, -Br, -CN, -NH 2 , -NHC 1-3 alkyl, methyl, ethyl, propyl, isopropyl, butyl, methoxy, ethoxy, propoxy or isopropoxy; C 1-3 alkyl substituted by halogen, -NH 2 , -OH, C 1-3 alkyl or C 1-3 alkoxyl; or C 1-3 alkoxyl substituted by halogen, -NH 2 , -OH, C 1-3 alkyl or C 1-3 alkoxyl.
  • the substituent may be selected from -F, -Cl, -Br, -OH, -NO 2 , -NH 2 , -NH(C 1-6 alkyl), -N(C 1-6 alkyl) 2 , -CN, C 1-6 alkyl, C 1-4 alkoxyl, C 1-4 alkoxyl-C 1-6 alkyl, C 3-8 cycloalkyl-C 1-8 alkyl, C 1-6 alkyl carbonyl, C 1-6 alkoxyl carbonyl, C 1-6 alkyl thiol, -S(O) 2 N(C 1-6 alkyl) 2 , -S(O) 2 C 1-6 alkyl, -N(C 1-6 alkyl)S(O) 2 N(C 1-6 alkyl) 2 , -S(O)N(C 1-6 alkyl) 2 , -S(O)(C 1-6 alkyl), -N(C 1-6 alkyl)S(O)N
  • the substituent may be selected from -F, -Cl, -Br, -OH, -NO 2 , -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CN, C 1-3 alkyl, C 1-3 alkoxyl, C 1-3 alkyl carbonyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, phenyl, naphthyl, anthracyl, phenanthryl, fluorenyl, thiophenyl, imidazolyl, pyrazolyl,
  • the substituent may be selected from -F, -Cl, -Br, -OH, -NO 2 , -NH 2 , -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 , -CN, methyl, ethyl, propyl, isopropyl, butyl, methoxy, ethoxy, propoxy, isopropoxy or phenyl.
  • the ring C may be selected from any of the following groups: wherein:
  • the ring C may be selected from any of the following groups: wherein:
  • the ring C may be selected from any of the following groups:
  • the ring A may be selected from substituted or unsubstiuted C 4-6 cyclic hydrocarbyl, substituted or unsubstiuted 4 to 6-membered heterocyclyl, substituted or unsubstiuted C 5-6 aryl, or substituted or unsubstiuted 5 to 6-membered heteroaryl, wherein the heterocyclyl or heteroaryl comprises 1-3 N atoms.
  • the ring A may be selected from any of the following groups:
  • the ring A may be selected from any of the following groups:
  • the compound may have a structure selected from:
  • the isotopic substitution of the isotope-substituted derivative of the compound herein may relate to atom comprises but not limited to hydrogen, carbon, nitrogen, oxygen, fluorine, phosphorus, chlorine or iodine; and preferably is 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl or 125 I.
  • the disease is cancer, preferably is Noonan Syndrome, Leopard Syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myeloid leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, head cancer, neuroblastoma, squamous cell carcinoma of head and neck, gastric cancer, anaplastic large cell lymphoma or glioblastoma.
  • composition comprising:
  • a method for inhibiting SHP2 acitivity comprising the following step: administering to a subject in need thereof an effective amount of a compound of Formula I of the disclosure or a pharmaceutically acceptable salt thereof, or administering to a subject in need thereof an effective amount of a pharmaceutical composition of the disclosure.
  • novel allosteric inhibitor compounds of Formula I which can achieve the purpose of inhibiting SHP2 activity by binding to the non-catalytic region of SHP2 and "locking" the self-inhibiting state with weak SHP2 activity.
  • the compounds of the disclosure exhibit very good biological activity and druggability, and have very good drug development prospects; they have an inhibitory effect on SHP2 at very low concentrations (as low as ⁇ 100nM/L), and the inhibitory activity is quite excellent, so they can be used to treat SHP2-related diseases or conditions, such as tumors.
  • the inventors completed the invention.
  • reactions and purifications may be carried out according to the manufacturer's instructions for use of kit, or in a manner well known in the art or according to the description of the disclosure.
  • the above techiques and methods can be implemented in accordance with the conventional methods well known in the art according to the descriptions in the multiple general and more specific literatures cited and discussed in the present description.
  • groups and substituents may be selected by a person skilled in the art to provide stable structural parts and compounds.
  • substituent When a substituent is described by a conventional formula written from left to right, the substituent also includes the chemically equivalent substituent obtained when the structural formula is written from right to left.
  • -CH 2 O- is equal to -OCH 2 -.
  • C1-C6 alkyl refers to an alkyl as defined below having a total of 1 to 6 carbon atoms.
  • the total number of carbon atoms in the simplified symbols does not comprise carbon atoms that may exist in the substituents of the group.
  • halogen refers to fluorine, chlorine, bromine or iodien.
  • Hydrophill refers to -OH group.
  • Hydroxyl alkyl refers to an alkyl as defined below substituted by hydroxyl (-OH).
  • Niryl refers to -NO 2 .
  • Cyano refers to -CN.
  • Amino refers to -NH 2 .
  • substituted amino refers to an amino substituted by one or two of the alkyl, alkyl carbonyl, aryl alkyl, heteroaryl alkyl as defined below, for example, substituted amino may be monoalkyl amino, dialkyl amino, alkyl acylamino, aryl alkyl amino, heteroaryl alkyl.
  • Carboxyl refers to -COOH.
  • alkyl refers to a fully saturated straight or branched hydrocarbon chain group, consisting only of carbon atoms and hydrogen atoms, for example, comprising 1 to 12 (preferably 1 to 8, more preferably 1 to 6) carbon atoms, and connected to the rest of the molecule by a single bond.
  • alkyl includes, but is not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-amyl, 2-methylbutyl, 2,2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl and decyl, etc.
  • the term "alkyl” refers to an alkyl group containing 1 to 8 carbon atoms.
  • alkenyl refers to a straight or branched hydrocarbon chain group, consisting only of carbon atoms and hydrogen atoms, for example, comprising 2 to 20 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, comprsing at least one double bond, and connected to the rest of the molecule by a single bond, Examples, include but are not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1-enyl, pent-1,4-dienyl, etc.
  • cyclic hydrocarbyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbyl (such as alkyl, alkenyl or alkynyl) consisting only of carbon atoms and hydrogen atoms, which may comprise fused ring system, bridged ring system or spiro ring system, comprise 3 to 15 carbon atoms, preferably comprise 3 to 10 carbon atoms, more preferably comprise 3 to 8 carbon atoms, for example, comprise 3, 4, 5, 6, 7 or 8 carbon atoms, and which is saturated or unsaturated and may be connected to the rest of the molecule via any suitable carbon atom by a single bond.
  • cyclic hydrocarbyl refers to a stable non-aromatic monocyclic or polycyclic hydrocarbyl (such as alkyl, alkenyl or alkynyl) consisting only of carbon atoms and hydrogen atoms, which may comprise fused ring system, bridged ring system or spiro ring system, comprise 3 to 15 carbon
  • cyclic hydrocarbyl include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2,3-dihydroindenyl, 1,2,3,4-tetrahydro-naphthyl, 5,6,7,8-tetrahydro-naphthyl, 8,9-dihydro-7H-benzocycloheptene-6-yl, 6,7,8,9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9,10-hexahydro-benzocyclooctenyl, fluor
  • heterocyclyl refers to a stable 3 to 20-membered non-aromatic cyclic group consisting of 2 to14 carbon atoms (for example, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen or sulfur.
  • a heterocyclyl may be a monocyclic ring system, a dicyclic ring system, a tricyclic ring system or a ring system with more rings, and may comprise fused ring system, bridged ring system or spiro ring system; the nitrogen, phosphorus or sulfur atoms in the heterocyclyl may be optionally oxidized; the nitrogen atoms in the heterocyclyl may be optionally quaternized; and the heterocyclyl may be partially or fully saturated.
  • the heterocyclyl may be connected to the rest of the molecule via a carbon atom or a heteroatom by a single bond.
  • heterocyclyl In a heterocyclyl containing fused ring, one or more rings may be aryl or heteroaryl as defined below, provided that the connection point between the group and the rest of the molecule is a non-aromatic ring atom.
  • heterocyclyl is preferably a stable 4 to 11-membered non-aromatic monocyclic, dicyclic, bridged or spiro ring group comprising 1 to 3 heteroatoms selected from nitrogen, oxygen or sulfur, more preferably a stable 4 to 8-membered non-aromatic monocyclic, dicyclic, bridged or spiro ring group comprising 1 to 3 heteroatoms selected from nitrogen, oxygen or sulfur.
  • heterocyclyls include but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2,7-diaza-spiro[3.5]nonane-7-yl, 2-oxa-6-aza-spiro[3.3]heptane-6-yl, 2,5-diaza-bicyclo[2.2.1]heptane-2-yl, azacyclobutanyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, di
  • aryl refers to a conjugated hydrocarbon ring system group comprising 6 to 18 carbon atoms (preferably comprising 6 to 10 carbon atoms, for example, 6, 7, 8, 9 or 10 carbon atoms).
  • aryl may be a monocyclic ring system, a dicyclic ring system, a tricyclic ring system or a ring system with more rings, and may be fused with the cyclic hydrocarbyl or heterocyclyl as defined above, provided that the aryl and the rest of the molecule are connected via an atom on the aromatic ring by a single bond.
  • Exemplary aryls include but are not limited to phenyl, naphthyl, anthracyl, phenanthrenyl, fluorenyl, 2,3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1,4-benzoxazin-3(4H)-one-7-yl, etc.
  • aryl alkyl refers to an alkyl as defined above which is substituted by an aryl as defined above.
  • heteroaryl refers to a 5 to 16-membered conjugated ring system group comprising 1 to 15 carbon atoms (preferably comprising 1 to 10 carbon atoms, for example, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen or sulfur.
  • a heteroaryl may be a monocyclic ring system, a dicyclic ring system, a tricyclic ring system or a ring system with more rings, and may be fused with the cycloalkyl or heterocyclyl, provided that the heteroaryl and the rest of the molecule are connected via an atom on the aromatic ring by a single bond.
  • the nitrogen, carbon or sulfur atoms in the heteroaryl may be optionally oxidized; the nitrogen atoms in the heteroaryl may be optionally quaternized.
  • heteroaryl is preferably a stable 5 to 12-membered aromatic group comprising 1 to 5 heteroatoms selected from nitrogen, oxygen or sulfur, and is more preferably a stable 5 to 10-membered aromatic group comprising 1 to 4 heteroatoms selected from nitrogen, oxygen or sulfur, or a 5 to 6-membered aromatic group comprising 1 to 3 heteroatoms selected from nitrogen, oxygen or sulfur.
  • heteroaryls include but are not limited to thiophenyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furanyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindazolyl, purinyl, quinolinyl, isoquinolinyl, diazanaphthalenyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl,
  • heteroaryl alkyl refers to an alkyl as defined above which is substituted by heteroaryl as defined above.
  • optional or “optionally” means that the subsequently described event or condition may or may not occur, and such description includes both occurrence and non-occurrence of the event or condition.
  • optionally substituted aryl means an aryl being substituted or not being substituted, and such description includes both substituted aryl and unsubsititued aryl.
  • the "optional" substituent employed in the claims and description of the disclosure is selected from alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, cyano, nitro, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cyclic hydrocarbyl, or optional substituted heterocyclyl.
  • SHP2 refers to "Src Homolgy-2 phosphatase", also called SH-PTP2, SH-PT3, Syp, PTP1D, PTP2C, SAP-2 or PTPN11.
  • part refers to specific part or functional group in the molecular. Chemical part is generally considered to be chemical entity embedded or attached to molecule.
  • Stepisomer refers to a compound composed of the same atoms, bonded by the same bonds, but having different three-dimensional structures.
  • the present disclosure will cover various stereoisomers and mixtures thereof.
  • Tautomer refers to an isomer formed by the transfer of proton from one atom of a molecule to another atom of the same molecule. All tautomeric forms of a compound of the disclosure will also be included within the scope of the disclosure.
  • a compound of the disclosure or pharmaceutically acceptable salt thereof may comprise one or more chiral carbon atoms, and thus may produce enantiomers, diastereomers, and other stereoisomeric forms.
  • Each chiral carbon atom can be defined as (R)- or (s)- based on stereochemistry.
  • the disclosure is intended to include all possible isomers, as well as racemes and optically pure forms thereof.
  • a compound of the disclosure can be prepared by using raceme, diastereomer or enantiomer as raw material or intermediate.
  • the optically active isomers can be prepared by using chiral synthons or chiral reagents, or separated by conventional techniques, such as crystallization and chiral chromatography.
  • the disclosure also includes all suitable isotopic variations of the compounds of the present disclosure or pharmaceutically acceptable salts thereof.
  • Isotopic variations of the compounds of the present disclosure or pharmaceutically acceptable salts thereof are defined as those in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass often found in nature.
  • Isotopes that can be incorporated into the compounds of the present disclosure and their pharmaceutically acceptable salts thereof include but are not limited to H, C, N and O, for example, 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 35 S, 18 F, 36 Cl and 125 I.
  • Suitable isotopic variations of the compounds or pharmaceutically acceptable salts thereof of the present disclosure may be prepared by conventional techniques using appropriate isotopic variants of suitable reagents.
  • pharmaceutically acceptable salt includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
  • “Pharmaceutically acceptable acid addition salt” refers to a salt formed with an inorganic acid or an organic acid that can retain the biological effectiveness of the free base without other side effects.
  • Inorganic acid salts include but are not limited to hydrochloride, hydrobromide, sulfate, nitrate, phosphate, etc .
  • organic acid salts include but are not limited to formate, acetate, 2,2-dichloroacetate, trifluoroacetate, propionate, hexanoate, caprylate, caprate, undecylenate, glycolate, gluconate, lactate, sebacate, adipate, glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate, cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, methanesulfonate, benzenes
  • “Pharmaceutically acceptable base addition salts” refers to a salt formed with an inoragnic base or an organic base that can retain the biological effectiveness of the free acid without other side effects.
  • Salts derived from inorganic bases include but are not limited to sodium salt, potassium salt, lithium salt, ammonium salt, calcium salt, magnesium salt, iron salt, zinc salt, copper salt, manganese salt, aluminum salt, etc.
  • Preferred inorganic salts are ammonium salt, sodium salt, potassium salt, calcium satl and magnesium salt.
  • Salts derived from organic bases include but are not limited to the following salts: primary amines, secondary amines, and tertiary amines, substituted amines, including natural substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucosamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resin, etc.
  • Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, di
  • pharmaceutical compostion refers to a formulation of a compound of the present disclosure and a medium generally accepted in the art for delivering a biologically active compound to a mammal (e.g., human).
  • the medium includes a pharmaceutically acceptable carrier.
  • the purpose of the pharmaceutical composition is to promote the administration of living organisms, which facilitates the absorption of active ingredients and thus exerts biological activity.
  • pharmaceutically acceptable refers to a substance (such as a carrier or diluent) that does not affect the biological activity or properties of the compound of the present disclosure, and is relatively non-toxic, that is, the substance can be administered to an individual without causing adverse biological reactions or interacting with any components contained in the composition in an undesirable manner.
  • pharmaceutically acceptable excipient includes but is not limited to any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, corrigent, surfactant, wetting agent, dispersant, suspending agent, stabilizer, isotonic agent, solvent or emulsifier that is approved by relevant government regulatory agency to be acceptable for human or domestic animal use.
  • Tumor in the disclosure includes but is not limited to Noonan Syndrome, Leopard Syndrome, juvenile myelomonocytic leukemia, neuroblastoma, sarcoma, melanoma, articular chondroma, cholangioma, leukemia, breast cancer, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, small cell lung cancer, esophageal cancer, pancreatic cancer, lung squamous cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharynx cancer, brain cancer, bone cancer, kidney cancer, oral cancer/head cancer, neuroblastoma, squamous cell carcinoma of head and neck, anaplastic large cell lymphoma or glioblastoma and other diseases.
  • preventive include reducing the possiblity of the occurrence or deterioration of a disease or condition in a patient.
  • treatment and other similar synonyms as employed herein include the following meanings:
  • an "effective amount” for treatment is the amount of a composition comprising a compound as disclosed herein required to provide a clinically significant disease relief effect. Techniques such as dose escalation tests may be used to determine the effective amount suitable for any individual case.
  • taking refers to a method capable of delivering a compound or composition to a desired site for biological action. These methods include but are not limited to oral route, transduodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), local administration, and transrectal administration.
  • parenteral injection including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion
  • local administration and transrectal administration.
  • transrectal administration including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion
  • transrectal administration include those discussed in Goodman and Gilman, The Pharmacological Basis of Therapeutics, current ed.; Pergamon ; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa .
  • the compounds and compositions discussed herein are administered orally.
  • the terms “medicament combination”, “medicament co-administration”, “combined medication”, “administration of other treatments”, “administration of other therapeutic agents” and the like as employed herein refer to medical treatment obtained by mixing or combining more than one active ingredient, and include fixed and unfixed combinations of active ingredients.
  • the term “fixed combination” refers to the simultaneous administration of at least one compound described herein and at least one synergistic medicament to a patient in the form of a single entity or a single dosage form.
  • the term “unfixed combination” refers to simultaneous administration, co-administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient in the form of separate entities. These also apply to cocktail therapy, such as the administration of three or more active ingredients.
  • the functional group of an intermediate compound may need to be protected by an appropriate protecting group.
  • Such functional groups include hydroxyl, amino, sulfydryl and carboxylic acid.
  • Suitable hydroxyl-protecting groups include trialkylsilyl or diarylalkylsilyl (e.g. tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, etc.
  • Suitable protecting groups for amino, amidino and guanidino include tert-butoxycarbonyl, benzyloxycarbonyl, etc.
  • Suitable sulfydryl-protecting groups include -C(O)-R" (wherein R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl, etc.
  • Suitable carboxy-protecting groups include alkyl, aryl or aralkyl esters.
  • Protecting groups may be introduced and removed according to standard techniques known to one skilled in the art and as described herein. The use of protecting groups is described in detail in Greene, T. W. and P. G. M. Wuts, Protective Groups in Organi Synthesis, (1999), 4th Ed., Wiley . Protecting groups may also be polymer resins.
  • the compound of Formula I provided by the present disclosure may be prepared by the following method: performing nucleophilic substitution reaction bewteen Formula Ib and Formula Ic to obtain Formula Id; performing substitution reaction between Formula Id and Formula Ie to obtain Formula If; and deprotecting Formula If using acid to obtain the compound of Formula I:
  • Src Hommolgy-2 phosphatase is a protein tyrosine phosphatase encoded by the PTPN11 gene, which promotes various cell functions, including proliferation, differentiation, cell cycle maintenance, and migration.
  • SHP2 is involved in signaling via Ras-mitogen-activated protein kinase, JAK-STAT or phosphoinositide 3-kinase-AKT pathway.
  • SHP2 mediates the activation of receptor tyrosine kinases such as ErbB1and ErbB2, and Erk1 and Erk2 MAP kinases of c-Met.
  • SHP2 has two N-terminal Src Homolgy-2 domains (N-SH2 and C-SH2), a catalytic domain (PTP) and a C-terminal tail.
  • the two SH2 domains control the subcellular localization and functional regulation of SHP2.
  • the molecule exists in an inactive conformation, inhibiting its own activity via a binding network involving residues from N-SH2 and PTP domains.
  • SHP2 binds to specific tyrosine-phosphorylation sites, such as Gab1 and Gab2, on docking proteins via SHP2's SH2 domain. This causes conformational change, leading to SHP2 activation.
  • SHP2 is an important downstream signaling molecule for various receptor tyrosine kinases, including the receptors of platelet-derived growth factor (PDGF-R), fibroblast growth factor (FGF-R) and epidermal growth factor (EGF-R). SHP2 is also an important downstream signaling molecule that activates the mitogen-activated protein (MAP) kinase pathway, which can lead to cell transformation (a necessary condition for cancer development).
  • PDGF-R platelet-derived growth factor
  • FGF-R fibroblast growth factor
  • EGF-R epidermal growth factor
  • SHP2 is also an important downstream signaling molecule that activates the mitogen-activated protein (MAP) kinase pathway, which can lead to cell transformation (a necessary condition for cancer development).
  • MAP mitogen-activated protein
  • SHP2 significantly inhibits cell growth of lung cancer cell lines having SHP2 mutations or EML4/ALK translocation, as well as EGFR-amplified breast and esophageal cancer.
  • SHP2 is also a downstream gene of the activation of oncogenes in gastric cancer, anaplastic large cell lymphoma, and glioblastoma.
  • Noonan Syndrome (NS) and Leopard Syndrome (LS)-PTPN1l mutation causes LS (multiple pigmented nevus syndrome, abnormal ECG conduction, distance between eyes too far, pulmonary valve stenosis, abnormal genitalia, growth retardation, sensorineural hearing loss) and NS (including congenital abnormalities of heart defect, craniofacial deformity and short stature).
  • LS multiple pigmented nevus syndrome, abnormal ECG conduction, distance between eyes too far, pulmonary valve stenosis, abnormal genitalia, growth retardation, sensorineural hearing loss
  • NS including congenital abnormalities of heart defect, craniofacial deformity and short stature.
  • Y62D and Y63C in the protein are the most common mutations. These two mutations affect the non-catalytic activity conformation of SHP2, but do not interfere with the binding of phosphatase and its phosphorylated signaling ligand.
  • JMML Juvenile myelomonocytic leukemia
  • SHP2 Juvenile myelomonocytic leukemia
  • MPD childhood myelodysplastic disease
  • Acute myeloid leukemia - PTPN1l mutations have been identified in about 10% of pediatric acute leukemias such as myelodysplastic syndrome (MDS), about 7% of B-cell acute lymphoblastic leukemia (B-ALL) and about 4% of acute myeloid leukemia (AML).
  • MDS myelodysplastic syndrome
  • B-ALL B-cell acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • NS and leukemia mutations cause changes of the amino acids at the interface formed by the N-SH2 and PTP domains in the self-inhibiting SHP2 conformation, disrupt inhibitory intramolecular interactions, and result in hyperactivity of the catalytic domain.
  • SHP2 acts as a positive regulator in receptor tyrosine kinase (RTK) signaling.
  • Cancers containing RTK changes include esophageal cancer, breast cancer, lung cancer, colon cancer, gastric cancer, glioma, and head and neck cancer.
  • Esophageal cancer (or esophagus cancer) is a malignant disease of the esophagus.
  • Esophageal cancer There are many subtypes of esophageal cancer, mainly squamous cell carcinoma ( ⁇ 50%) and adenocarcinoma.
  • squamous cell carcinoma ⁇ 50%)
  • adenocarcinoma adenocarcinoma
  • RTK expression esophageal adenocarcinoma and squamous cell carcinoma. Therefore, the SHP2 inhibitor of the disclosure can be used for innovative treatment strategies.
  • TNBC triple-negative breast cancer
  • EGFR Epidermal growth factor receptor I
  • SHP2 may be a promising treatment for breast cancer.
  • Lung cancer - NSCLC is currently an important cause of cancer-related mortality. It accounts for about 85% of lung cancer (mainly adenocarcinoma and squamous cell carcinoma). Although cytotoxic chemotherapy is still an important part of treatment, targeted therapies based on genetic changes (such as EGFR and ALK) in tumors are more likely to benefit from targeted therapy.
  • Colon cancer About 30% to 50% of colorectal tumors are known to have mutated (abnormal) KRAS, and BRAF mutations occur in 10% to 15% of colorectal cancers. For a subgroup of patients whose colorectal tumors have been shown to overexpress EGFR, these patients present a favorable clinical response to anti-EGFR therapy.
  • Gastric cancer is one of the most popular types of cancer.
  • the abnormal expression of tyrosine kinase (as reflected by abnormal tyrosine phosphorylation in gastric cancer cells) is known in the art.
  • Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system, accounting for about 8% of childhood cancers. Genomic changes of anaplastic lymphoma kinase (ALK) gene have been proposed to promote the pathogenesis of neuroblastoma.
  • ALK anaplastic lymphoma kinase
  • Squamous cell carcinoma of the head and neck High levels of EGFR expression are associated with poor prognosis and resistance to radiotherapy in a variety of cancers, most commonly squamous cell carcinoma of the head and neck (SCCHN).
  • Blocking of EGFR signal leads to the inhibition of receptor stimulation and the decrease of cell proliferation, invasion and metastasis. Therefore, EGFR is the best target of new anticancer therapy in SCCHN
  • the present disclosure relates to compounds capable of inhibiting SHP2 activity.
  • the disclosure also provides a preparation method of the compound of the disclosure and a pharmaceutical preparation containing the compound.
  • Another aspect of the disclosure relates to a method of treating a SHP2-mediated disease or condition, which comprises a step of administering to a patient in need thereof a therapeutically effective amount of the compound of Formula I of the disclosure.
  • the disclosure relates to the method as described above, wherein the SHP2-mediated disease or condition is seleted from but not limited to the following cancers: JMML, AML, MDS, B-ALL, neuroblastoma, esophageal cancer, breast cancer, lung cancer, colon cancer, stomach cancer, head and neck cancer.
  • the compound of the disclosure also may be used for treating other diseases or conditions related to abnormal SHP2 activity. Therefore, as a preferred embodiment, the disclosure relates to a method for treating a disease or condition selected from: NS, LS, JMML, AML, MDS, B-ALL, neuroblastoma, esophageal cancer, breast cancer, lung cancer, colon cancer, stomach cancer, head and neck cancer.
  • a disease or condition selected from: NS, LS, JMML, AML, MDS, B-ALL, neuroblastoma, esophageal cancer, breast cancer, lung cancer, colon cancer, stomach cancer, head and neck cancer.
  • the SHP2 inhibitors of the present disclosure may be combined with another pharmacologically active compound or with two or more other pharmacologically active compounds, especially in the treatment of cancer.
  • the compound of Formula (I) of the present disclosure or a pharmaceutically acceptable salt thereof may be administered simultaneously, sequentially, or separately in combination with one or more substances selected from: chemotherapeutic agents, such as mitotic inhibitors, such as taxane, vinca alkaloids, paclitaxel, docetaxel, vincristine, vinblastine, vinorelbine or vinflunine, and other anticancer agents, such as cisplatin, 5-fluorouracil or 5-fluoro-2-4(1H,3H)-pyrimidinedione (5FU), flutamide or gemcitabine.
  • mitotic inhibitors such as taxane, vinca alkaloids, paclitaxel, docetaxel, vincristine, vinblastine, vinorelbine or vinflunine
  • other anticancer agents such as cis
  • Some combinations can provide significant benefits in therapy, including synergistic activity.
  • the disclosure relates to the method as described above, wherein the compound is administrated parenterally.
  • the disclosure relates to the method as described above, wherein thecompound is administered intramuscularly, intravenously, subcutaneously, orally, pulmonaryly, intrathecally, topically or intranasally.
  • the disclosure relates to the method as described above, wherein the compound is administered systemically.
  • the disclosure relates to the method as described above, wherein the patient is a mammal.
  • the disclosure relates to the method as described above, wherein the patient is a primate.
  • the disclosure relates to the method as described above, wherein the patient is a human.
  • the disclosure relates to the method of treating a SHP2-mediated disease or condition, wherein the method comprises the following step: administering to a patient in need thereof a combination of a therapeutically effective amount of chemotherapeutic agent and a therapeutically effective amount of the compound of Formula I of the disclosure.
  • the starting materials used in the following examples can be purchased from chemical distributors such as Aldrich, TCI, Alfa Aesar, Bide, Energy, etc., or can be synthesized by known methods.
  • the ice bath refers to -5 °C to 0 °C
  • the room temperature refers to 10 °C to 30 °C
  • the reflux temperature generally refers to the reflux temperature of the solvent under normal pressure.
  • Overnight reaction generally refers to a time of 8-15 hours. In the following examples, operations without a specific operating temperature are all carried out at room temperature.
  • the separation and purification of the intermediate and final products are performed by normal phase or reversed phase chromatography column separation or other suitable methods.
  • Normal phase flash chromatography columns use ethyl acetate and n-hexane or methanol and methylene chloride as mobile phases.
  • Reversed phase preparative high-pressure liquid chromatography (HPLC) uses a C18 column and UV at 214 nm and 254 nm with mobile phases A (water and 0.1% formic acid) and B (acetonitrile), or mobile phases A (water and 0.1 % Ammonium bicarbonate) and B (acetonitrile).
  • Step 1 1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester (1.51g, 5mmol), tetraethyl titanate (6.84g, 30mmol) and (R)-(+)-tert-butylsulfinamide (2.41g, 20mmol) were successively added to a dry 100mL single-necked flask, and the mixture was stirred under heating and reflux for 15 hours. After the reaction system was cooled to room temperature, saturated brine (60 mL) was added to the reaction residue, after which the resulting mixture was stirred for 15 minutes and then filtered through diatomite.
  • saturated brine 60 mL
  • Step 2 (R,Z)- 1-((tert-butylsulfinyl)imino)- 1,3 -dihydrospiro[indene-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A1-1 , 0.802g, 2mmol) and THF (10mL) were successively added to a dry 100mL single-necked flask, the mixture was cooled to 0°C, and then lithium borohydride (66mg, 3mmol) was added. The resulting mixture was then stirred for 1 hour. Methanol was slowly added to quench excess borohydride. The reaction solution was filtered, and concentrated, and volatiles were removed under reduced pressure.
  • Step 3 (S)-1 -(((R)-tert-butylsulfinyl)amino)-1,3 -dihydrospiro[indene-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A1-2 , 0.406g, 1mmol), dichloromethane (5mL), and TFA (1mL) were successively added to a dry 50mL single-necked flask, and the resulting mixture was stirred at room temperature for 1 hour. Na 2 CO 3 saturated aqueous solution was added until pH reached 7, and the aqueous mixture was extracted with DCM (3 ⁇ 30mL).
  • Step 1 4-cyanopiperidine-1-carboxylic acid tert-butyl ester (1.05g, 5mmol) and THF (20mL) were successively added to a dry 100mL flask. Under the protection of nitrogen, the mixture was cooled to -78 °C, and then 2M of LDA(3.3mL, 6.5mmol) was slowly added to the reaction mixture. The reaction mixture was allowed toreact for 1 hour, and then 3-bromo-2-(bromomethyl)pyridine (1.24g, 5mmol) was added thereto, and then the reaction mixture was allowed to continue to react for 2 hours.
  • Step 3 5-oxo-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A2-2 , 0.302g, 1mmol), tetraethyl titanate (1.37g, 6mmol) and (R)-(+)-tert-butylsulfinamide (0.480g, 4mmol) were successively added to a dry 100mL single-necked flask and the mixture was stirred under heating and reflux for 15 hours.
  • Step 4 (R,Z)-5-((tert-butylsulfinyl)imino)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A2-3 , 0.20g, 0.491mmol) and THF (50mL) were successively added to a dry 100mL single-necked flask, the mixture was cooled to 0°C, and then lithium borohydride (0.018g, 0.737mmol) was added. The resulting mixture continued to react with stirring for 1 hour. Methanol was slowly added to quench excess borohydride.
  • Step 5 (S)-5-((R)-tert-butylsulfonamido)-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A2-4 , 0.100g, 0.245mmol), dichloromethane (5mL), and TFA(1mL) were successively added to a dry 50mL single-necked flask, and the resulting mixture was reacted with stirring at room temperature for 1 hour. Na 2 CO 3 saturated aqueous solution was added until pH reached 7, and the aqueous mixture was extracted with DCM (3 ⁇ 30mL).
  • Step 1 A6-1 (11.1g, 60mmol) and NaBH 4 (2.51g, 66mmol) were successively added to 300mL of THF and 60mL of H 2 O in a 1L flask, and the mixture was reacted at 20 °C for 2 hours. After the raw materials were confirmed to be completely reacted by spot plate detection, the reaction mixture was quenched with saturated NH 4 Cl solution, diluted with water, and extracted with ethyl acetate. The combined organic phase was washed with saturated NaCl solution, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain 2-bromo-pyridin-3-ylmethanol ( A6-2 , 11.2g, yield: 100%) as a white solid.
  • Step 2 A6-2 (6.73g, 36mmol) and Ms 2 O (6.96g,g, 40mmol) were successively added to 120mL of dichloromethane in a dry 250mL single-necked flask, then the mixture was cooled to 0°C, then TEA (5.45g, 54mmol) was slowly added, and then the mixture was heated to room temperature and stirred for 3 hours. After the reaction was completed, the reaction solution was washed with water. The organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 3 4-ethoxycarbonylpiperidine-1-carboxylic acid tert-butyl ester (9.3g, 36.2mmol) and THF (133mL) were successively added to a dry 500mL three-necked flask. Under the protection of nitrogen, the mixture was cooled to -70 °C, and then 2M of LDA (21.1mL, 42.3mmol) was slowly added to the reaction mixture.
  • reaction mixture was allowed to react for 1 hour, then methylsulfonic acid (2-bromopyridin-3-yl)methyl ester (A6-3, 8.0 g, 30.2 mmol) dissolved in 65 mL of THF was added thereto, then the reaction mixture was allowed to continue to react for 0.5 hours, and then the reaction mixture was slowly heated to room temperature and stirred for 1hour. After the reaction was completed, the reaction mixture was quenched by saturated brine and extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • the obtained residue was purified by silica gel chromatography (8 to 12% gradient of ethyl acetate/ dichloromethane) to obtain 4-ethoxycarbonyl-4-(2-bromo-3-pyridyl)methyl-piperidine-1-carboxylic acid tert-butyl ester ( A6-4 , 10.5g, yield: 81%) as a colorless oily matter.
  • Step 4 4-ethoxycarbonyl-4-(2-bromo-3-pyridyl)methyl-piperidine-1-carboxylic acid tert-butyl ester ( A6-4 , 7.85g, 18.4mmol) and THF (120mL) were successively added to a dry 250mL three-necked flask. Under the protection of nitrogen, the mixture was cooled to -70 °C, and then 2.5 M of n-butyllithium (11mL, 27.6mmol) was slowly added to the reaction mixture. The reaction mixture was allowed to react for 1.5 hours.
  • the obtained residue was purified by silica gel chromatography (0 to 60% gradient of ethyl acetate/petroleum ether) to obtain 7-oxo-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A6-5 , 1.4g, yield: 25%) as a light brown solid.
  • Step 5 Intermediate (R,Z)-7-((tert-butylsulfinyl)imino)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1' -carboxylic acid tert-butyl ester ( A6-6 , 1.46g, yield: 68%) was synthesized according to the synthesis protocol of intermediate A2-3, using intermediate A6-5 instead of intermediate A2-2.
  • Step 6 (R,Z)-7-((tert-butylsulfinyl)imino)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1' -carboxylic acid tert-butyl ester ( A6-6 , 530mg, 1.31mmol) and THF (10mL) were successively added to a dry 50mL three-necked flask. Under the protection of nitrogen, the mixture was cooled to -70 °C, and then 1.5 M of diisobutylaluminum hydride in toluene (1.3 mL, 1.95 mmol) was slowly added to the reaction mixture.
  • reaction mixture was allowed to react for 0.5 hours, and then the mixture was slowly heated to room temperature, quenched with saturated potassium sodium tartrate aqueous solution and stirred for 0.5 hours.
  • the reaction mixture was extracted with ethyl acetate. The organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • Step 7 (S)-7-((R)-tert-butylsulfonamido)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A6-7 , 312mg, 0.766mmol), MeOH (5mL), and 4M of HCl/1,4-dioxane solution (3.83mL, 15.3mmol) were successively added to a dry 25mL single-necked flask, and then the mixture was heated to 50 °C and reacted for 8 hour to obtain a white suspension.
  • Step 1 Intermediate 4-ethoxycarbonyl-4-(4-fluorobenzyl)-pyridine-1-carboxylic acid tert-butyl ester A7-1 was synthesized according to the synthesis protocol of intermediate A2, using raw material 4-fluorobenzyl bromide instead of raw material 3-bromo-2(bromomethyl)pyridine.
  • Step 2 4-ethoxycarbonyl-4-(4-fluorobenzyl)-pyridin-1-carboxylic acid tert-butyl ester ( A7-1 , 3.40g, 9.30mmol) and sodium hydroxide (1.86g, 46.5mmol) were successively added to 20mL of methyl alcohal and 20mL of water in a 100mL singel-necked flask. The reaction solution was reacted at 70 °C for 17 hours. The reaction mixture was cool to room temperature, and then concentrated under reduced pressure to get rid of the volatiles.
  • Step 3 1-tert-butoxycarbonyl-4-(4-fluorobenzyl)-pyridine-4-carboxylic acid ( A7-2 , 2.0g, 5.93mmol) and PPA (15mL) were successively added to a dry 50mL single-necked flask. The reaction solution was reacted at 120 °C for 2 hours. The reaction solution was poured into ice-water mixture (50mL) while it was still hot, and NaOH solid was added to adjust pH to 10. Then, Boc 2 O (1.94g, 8.90mmol) was added to the resulting mixture, and the reaction solution was stirred at 20 °C for 1 hour. The reaction solution was extracted 3 times with ehtyl acetate (80mL).
  • Step 4 Intermediate (R,Z)-1-((tert-butylsulfinyl)imino)-6-fluoro-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine]-1' -formate A7-4 was synthesized according to the synthesis protocol of intermediate A2, using intermediate A7-3 instead of intermediate A2-2.
  • Step 5 (R,Z)-1-((tert-butylsulfinyl)imino)-6-fluoro-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine]-1' -formate ( A7-4 ,1.59g, 3.76mmol), tetrahydrofuran/water (98:2, 32mL) and sodium borohydride (427mg, 11.3mmol) were successively added to a round-bottom flask at -50 °C, and the reaction solution was heated to 20 °C in 3 hours undering stirring. The reaction was confirmed to be complete by TLC spot plate.
  • Step 6 Intermediate (S)-6-fluoro-1,3-dihydrospiro[indene-2,4'-piperidine]-1-amine A7 was synthesized according to the synthesis protocol of intermediate A3, using intermediate A7-5 instead of intermediate A1-2.
  • Step 1 4-bromonicotinicaldehyde hydrobromide salt ( A8-1 , 2.5g, 9.36mmol) was dissolved in methyl alcohal (50mL), sodium borohydride (0.72g, 18.93mmol, 2.0eq) was added in batches in an ice bath, and the mixture was reacted at 0 °C for 1 hour. After the reaction was completed, saturated ammonium chloride aqueous solution (50mL) was added in an ice bath to quench the reaction, and the reaction mixture was extracted with ethyl acetate (100mL ⁇ 2). The organic phases were combined, washed with saturated sodium chloride aqueous solution.
  • Step 2 (4-bromopyridin-3-yl)methanol ( A8-2 , 1.7g, 9.04mmol) was dissolved in dichloromethane (100mL), and triethylamine (2.30g, 22.7mmol) was added in an ice bath. Under the protection of nitrogen, methanesulfonic anhydride (1.95g, 11.2mmol) was added in batches, the mixture was reacted at 0 °C for 2 hours, and then the reaction was completed. Saturated sodium chloride (50mL) was added to quench the reaction in an ice bath, and the organic phase was seperated and washed with saturated sodium chloride aqueous solution.
  • Step 3 4-cyanopiperidine-1-carboxylic acid tert-butyl ester (1.61g, 7.67mmol) was dissolved in andyrous tetrahydrofuran (60mL). In a dry ice acetone bath (-78 °C) and the protection of nitrogen, LDA(4.6mL, 9.2mmol) was slowly added dropwise, the mixture was reacted for 0.5 hours while the temperature was maintained, and a lot of white solid precipitated from the reaction solution.
  • Step 4 4-((4-bromopyridin-3-yl)methyl)-4-cyanopiperidine -1-carboxylic acid tert-butyl ester ( A8-4 , 1.5g, 3.95mmol) was dissolved in DMAc/H 2 O (100mL/10mL), DIPEA (1.34g, 15.8mmol) and Pd(AmPhos) 2 Cl 2 (142mg, 0.20mmol) were added, the atmosphere in the reaction system was replaced with nitrogen three times, the mixture was reacted at 130 °C for 2 hours, and then the reaction was completed. Water (50mL) was added, and the mixture was extracted with ethyl acetate (100mL ⁇ 3).
  • Step 5 5-oxo-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A8-5 , 750mg, 2.48mmol) was dissolved in THF (10mL), and (R)-(+)-tert-butylsulfinamide (390mg, 3.22mmol) and tetraethyl titanate (10mL) were added. Under the protection of nitrogen, the mixture was heated to 90 °C and refluxed for 18 hours.
  • reaction mixture was cooled to room temperature and diluted with ethyl acetate (50mL), saturated brine (10mL) was added, and white solid precipitated.
  • the mixture was filtered, and the filter cake was washed with ethyl acetate.
  • the filtrate was washed with saturated brine, and the organic phase was dried over anhydrous sodium sulfate, filtered and concentrated.
  • Step 6 (R,Z)-5-((tert-butylsulfinyl)imino)-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A8-6 , 800mg, 1.98mmol) was dissolved in THF (50mL), and the mixture was cooled to -78 °C under the protection of nitrogen. 1.5M of DIBAL-H (2mL, 3mmol) was slowly dripped into the reaction solution. After dripping was completed, the mixture was reacted at -78 °C for 1 hour, and then the reaction was completed.
  • Step 7 (S)-5-(((R)-tert-butylsulfinyl)amino)-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A8-7 , 680mg, 1.67mmol) was dissolved in MeOH (50mL), the mixture was cooled to 0°C, 4 M of hydrochloric acid dioxane solution (10mL, 40mmol) was added dropwise, the resulting mixture was react at room temperature for 3hours, and then the reaction was completed.
  • Step 1 2-bromo-6-hydroxybenzaldehyde (5g, 24.9mmol) was dissolved in DMF (100mL), anhydrous potassium carbonate (6.88g, 49.8mmol) and 4-methoxybenzyl bromide (5.26g, 26.1mmol) was added, the mixture was reacted at room temperature for 18 hours under nitrogen protection, and then the reaction was completed.
  • the reaction solution was poured into ice water, and extracted with ethyl acetate (250mL) twice. The organic phases were combined and washed with saturated brine. The organic phase was separated, dried over anhydrous sodium sulfate, filtered and concentrated.
  • Step 2 2-bromo-6-((4-methoxybenzyl)oxy)benzaldehyde ( A9-1 , 8g, 24.9mmol) was dissolved in ethanol (100mL) and the mixture was cooled to 0 °C in an ice bath. Sodium borohydride (942mg, 24.9mmol) was carefully added in multiple batches. The mixture was reacted at 0 °C for 0.5 hours, and then the reaction was completed. The reaction solution was poured into ice water and extracted twice with ethyl acetate (200mL).
  • Step 3 (2-bromo-6-((4-methoxybenzyl)oxy)phenyl)methanol ( A9-2 , 7.3g, 22.6mmol) was dissolved in dichloromethane (200mL), and the mixture was cooled in an ice bath to 0 °C. Carbon tetrabromide (11.2g, 33.9mmol) and triphenylphosphine (8.88g, 33.9mmol) were added under nitrogen protection. The mixture was reacted at 0 °C for 5 hours, and then the reaction was completed. The reaction solution was poured into ice water and extracted twice with ethyl acetate (200mL).
  • Step 4 4-cyanopiperidine-1-carboxylic acid tert-butyl ester (2.6 g, 12.4 mmol) was dissolved in anhydrous THF (60 mL) and the mixture was cooled to -78 °C. 2M of LDA (7.5mL, 14.9mmol) was slowly added dropwise. The mixture was reacted at -78 °C for 0.5 hours, and then a solution of 1-bromo-2-(bromomethyl)-3-((4-methoxybenzyl)oxy)benzene ( A9-3 , 4g, 10.4mmol) in anhydrous THF (40 mL) was added dropwise.
  • Step 5 4-(2-bromo-6-((4-methoxybenzyl)oxy)benzyl)-4-cyanopiperidine-1-carboxylic acid tert-butyl ester ( A9-4 , 2.6g, 5.0mmol ) was dissolved in DMAc (100mL), H 2 O (10mL), DIPEA(3.225g, 25.0mmol) and PdCl 2 (AmPhos) 2 (354mg, 0.5mmol) were added. The mixture was reacted at 130 °C under argon protection for 18 hours, and then the reaction was completed. After cooled to room temperature, the reaction solution was poured into ice water and extracted twice with ethyl acetate (300 mL).
  • Step 6 4-((4-methoxybenzyl)oxy)-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A9-5 , 1.8g, 4.12mmol) was dissolved in THF (20mL), (R)-2-methylpropane-2-sulfinamide (748mg, 6.18mmol) and tetraethyl titanate (50mL) were added. The reaction mitxture was heated to 100 °C and refluxed for 18 hours under nitrogen protection.
  • Step 7 (R,Z)-1-((tert-butylsulfinyl)imino)-4-((4-methoxybenzyl)oxy)-1,3-dihydrospiro[indene-2,4'-pi peridine]-1'-carboxylic acid tert-butyl ester ( A9-6 , 1.7g, 3.15mmol) was dissolved in THF (100mL) and the mixture was cooled to -78 °C under nitrogen protection. 1.5M of DIBAL-H (3.15mL, 4.7mmol) was slowly added dropwise to the reaction solution. After the addition was completed, the reaction solution was reacted at -78 °C for 1 hour until the reaction was completed.
  • Step 8 (S)-1-(((R)-tert-butylsulfinyl)amino)-4-((4-methoxybenzyl)oxy)-1,3-dihydrospiro[indene-2,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A9-7 , 1.5g, 2.77mmol) was dissolved in MeOH (50mL), the mixture was cooled to 0 °C, and 4 M of hydrochloric acid dioxane solution (10mL, 40mmol) was added dropwise. The mixture was reacted at room temperature for 3 hours, and then the reaction is completed.
  • Step 1 2-methyl-3-cyanobromobenzene (3.0g, 15.3mmol, 1.0eq), N-bromosuccinimide (2.72g, 15.3mmol, 1.0eq), dibenzoyl peroxide (371mg, 1.53mmol, 0.1eq) and carbon tetrachloride (40mL) was successively added to a 100mL round-bottom flask, and the mixture was stitted at 80°C for 16 hours. The reaction solution was concentrated under reduced pressure to obtain a residue.
  • Step 2 Tetrahydrofuran (30mL) and 4-cyanopiperidine-1-carboxylic acid tert-butyl ester (1.84g, 8.73mmol, 1.2eq) were successively added to a 100 mL round-bottom flask, the mixture was cooled to -78 °C, then 2.0 M of LDA(5.1 mL, 10.2mmol, 1.4eq) was added, and the mixture was stirred at -78°C for one hour.
  • Step 3 Tert-butyl-4-(2-bromo-6-cyanobenzyl)-4-cyanopiperidine-1-carboxylate ( A10-2 , 1.5g, 3.71mmol, 1.0eq), Pd(AmPhos) 2 Cl 2 (262mg, 0.37mmol. 0.1eq), diisopropylethylamine (2.4g, 18.5mmol, 5.0eq), N,N-dimethyl acetamide (30mL) and water (4mL) were successively added to a dry 100mL round-bottom three-necked flask. With stirring, the atmosphere in the reaction system was replaced with nitrogen three times, and then the reaction mixture was heated to 140 °C and reacted for 16 hours.
  • Step 4 Tert-butyl-4-cyano-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine]-1-formate ( A10-3 , 900mg, 2.76mmol), titanium tetraethoxylate (3.78g, 16.6mmol), (R)-(+)-tert-butylsulfinamide (401mg, 3.31mmol) and tetrahydrofuran (20mL) were successively added to a dry 100mL single-necked flask, and the mixture was stirred under heating and reflux for 16 hours.
  • Step 5 Tetrahydrofuran (15mL) and (R,Z)-1-((tert-butylsulfinyl)imide)-4-cyano-1,3-dihydrospiro[indene-2,4'-piperidine]-1'-carbo xylic acid tert-butyl ester ( A10-4 , 980mg, 2.28mmol, 1.0eq) was successively added to a dry 50 mL three-necked flask.
  • Diisobutyl aluminum hydride (6.8ml, 1.5 M in toluene, 10.3mmol, 4.5eq) was added dropwise at -78 °C and under nitrogen protection, and the mixture continued to be stirred for half an hour. Then the mixture was heated to 0 °C and continued to be stirred for half an hour. After the reaction was completed, the reaction mixture was quenched with potassium sodium tartrate (4 g dissolved in 20mL of water), stirred for half an hour, and extracted with ethyl acetate (30mL ⁇ 3). The obtained organic phase was washed with saturated brine (30mL), dred with anhydrous sodium sulfate, filtered, and concentrated under reduced pressure.
  • Step 1 2,2,6,6-tetramethylpiperidine (5.33g, 37.8mmol) was dissolved in 100mL of tetrahydrofuran, the mixture was cooled to -65 °C under argon protection in a dry ice acetone bath, n-butyl lithium (2.5M, 15.8mL, 39.5mmol) was added dropwise, then the temperature of the mixture was maintained at 0 °C for 30min and then lowed to -65 °C again, a mixture of 2-bromopyrazine (5.0g, 31.4mmol) and 10mL of tetrahydrofuran was added dropwise, the temperature was maintained for 30min, a mixture of DMF (5.75g, 78.6mmol) and 10 mL of tetrahydrofuran was added dropwise, the resulting mixture continued to be stirred for 2 hours, and then the reaction was complete.
  • 2-bromopyrazine 5.0g, 31.4mmol
  • Step 2 (3-bromopyridin-2-yl)methanol ( A11-1 , 4.33g, 22.9mmol) was dissolved in 80mL of ether, PBr 3 (6.83g, 25.2mmol) was added dropwise at 0 °C under the protection of nitrogen, then the mixture was heated to reflux at about 40 °C and reacted for 4 hours, and then the reaction was completed.
  • the reaction liquid was poured into an ice NaHCO 3 aqueous solution, extracted twice with dichloromethane, and washed once with saturated brine.
  • Step 3 N-Boc-4-piperidine ethyl formate (6.81g, 26.5mmol) was dissolved in 160mL of anhydrous tetrahydrofuran, the mixture was cooled to -65 °C in a dry ice acetone bath under the protection of argon, LDA(2.0 M, 16.8mL, 33.6mmol) was slowly added dropwise, and the mixture was reacted for 1 hour while the temperature was maintained.
  • Step 4 1-(tert-butyl)-4-ethyl-4-((3-bromopyrazin-2-yl)methyl)piperidine-1,4-dicarboxylate ( A11-3 , 5.8g, 13.5mmol) was dissolved in 300mL of tetrahydrofuran, the mixture was cooled to -65 °C in a dry ice acetone bath under the protection of argon, n-butyl lithium (2.5M, 8.2ml, 20.3mmol) was added dropwise, the mixture was naturally warmed up to -10°C and maintained for 3 hours, and then the reaction was complete.
  • n-butyl lithium 2.5M, 8.2ml, 20.3mmol
  • Step 5 5-oxo-5,7-dihydrospiro[cyclopenta[b]pyrazine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A11-4 , 2.23g, 7.35mmol) was dissolved in 350mL tetrahydrofuran, and (R)-2-methylpropane-2-sulfinamide (980mg, 8.09mmol) and tetraethyl titanate (21.8g, 95.6mmol) were added. Under nitrogen protection, the mixture was heated to 80 °C and refluxed for 10 hours. The reaction was completed.
  • the reaction mixture was cooled to room temperature, poured into 400mL of ice water, and extracted with 300mL of ethyl acetate for 3 times (the aqueous phase contained a large amount of white flocculent solid, and the organic phase was almost without solid).
  • the organic phase was washed once with 200mL of water and once with 200mL of saturated brine.
  • Step 6 (R,Z)-5-((tert-butylsulfinyl)imino)-5,7-dihydrospiro[cyclopenta[b]pyrazine-6,4'-piperidine]-1' -carboxylic acid tert-butyl ester ( A11-5 , 2.38g, 5.85mmol) was dissolved in 280mL of tetrahydrofuran, and the mixture was cooled to -65 °C in a dry ice acetone bath under the protection of argon. 1.5M of DIBAL-H (5.07mL, 7.61mmol) was slowly added dropwise, and the temperature of the mixture continued to be maintained for 2 hours, and then the reaction is completed.
  • DIBAL-H 5.07mL, 7.61mmol
  • Step 7 (S)-5-((R)-tert-butylsulfinyl)amino)-5,7-dihydrospiro[cyclopenta[b]pyrazine-6,4'-piperidine]-1'-carboxylic acid tert-butyl ester crude product ( A11-6 , 2.48g, 5.85mmol) was dissolved in 120mL of methanol, the mixture was cooled in ice water to about 0 °C under the protection of argon, 4 M of dioxane hydrochloride solution (18mL, 72mmol) was added dropwise, the mixture was reacted at room temperature for 2 hours, and then the reaction was completed.
  • Step 1 2-methyl-3-methoxybromobenzene (2.0g, 9.95mmol, 1.0eq), N-bromosuccinimide (1.77g, 9.95mmol, 1.0eq), dibenzoyl peroxide (241mg, 0.995mmol, 0.1eq) and carbon tetrachloride (40mL) was added into a 100mL round-bottom flask successively, the mixture was stirred at 80°C for 16 hours, and the reaction solution is concentrated under reduced pressure to obtain a residue.
  • Step 2 Tetrahydrofuran (30mL) and N-Boc-4-cyanopyridine (1.8g, 8.57mmol, 1.2eq) were added to a 100mL round-bottom flask successively, the mixture was cooled to -78°C, then 2.0 M of LDA (5mL, 10mmol, 1.4eq) was added and the mixture was stirred at -78°C for one hour. Then a solution of 1-bromo-2-(bromomethyl)-3-methoxybenzene ( A12-1 , 2.0g, 7.14mmol, 1.0eq) in tetrahydrofuran (15mL) was added and the mixture was stirred at -78°C for 0.5h. Then the low temperature bath was removed.
  • Step 3 N-Boc-4-(2-bromo-6-methoxybenzyl)-4-cyanopyridine ( A12-2 , 1.5g, 3.66mmol, 1.0eq), Pd(AmPhos) 2 Cl 2 (259mg, 0.37mmol, 0.1eq), diisopropylethylamine (2.37g, 18.3mmol, 5.0eq), N,N-dimethylacetamide (30mL) and water (4mL) were added to a dry 100mL round-bottom flask successively. Under stirring, the atmosphere in the reaction system was replaced with nitrogen three times, and then the reaction mixture was heated to 140 °C and reacted for 16 hours.
  • the reaction liquid is cooled to room temperature, diluted with ethyl acetate (100mL) and filtered under reduced pressure.
  • the filter cake was washed with ethyl acetate (20mL).
  • the obtained filtrate was washed with saturated brine (30ml ⁇ 3) for three times, dried over anhydrous sodium sulfate and filtered.
  • the filtrate was concentrated under reduced pressure to obtain a crude product.
  • the crude product was purified by silica gel chromatography to obtain N-Boc-4-methoxy-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine] ( A12-3 , 0.90g, yield: 74.1%) as a white solid.
  • Step 4 N-Boc-4-methoxy-1-oxo-1,3-dihydrospiro[indene-2,4'-piperidine] ( A12-3 , 900mg, 2.72mmol), tetraethoxy titanium (3.72g, 16.3mmol), (R)-(+)-tert-butylsulfinamide (395mg, 3.26mmol) and tetrahydrofuran (20mL) were added to a dry 100mL single-necked flask successively, and the mixture was stirred for 16 hours under heating and reflux.
  • the obtained residue was purified by silica gel chromatography (0-30% gradient of ethyl acetate: petroleum ether) to obtain N-Boc-(R,Z)-1-((tert-butylsulfinyl)imide)-4-methoxy-1,3-dihydrospiro[indene-2,4'-piperidine] ( A12-4 , 800mg, yield: 67.8%) as a white solid.
  • Step 5 Tetrahydrofuran (15mL) and N-Boc-(R,Z)-1-((tert-butylsulfinyl)imide)-4-methoxy-1,3-dihydrospiro[indene-2,4'-piperidine] ( A12-4 , 800mg, 1.84mmol, 1.0eq) were added to a dry 50mL three-necked flask successively. Diisobutyl aluminum hydride (5.52ml, 1.5M in toluene, 8.28mmol, 4.5eq) was added dropwise at -78°C under nitrogen protection, and the mixture continued to stirred for half an hour.
  • Step 1 1-tert-butyl 4-ethylpiperidine-1,4-dicarboxylate (10.7g, 41.6mmol, 1.2eq) was dissolved in THF (50mL), LDA(2M, 9.8mL, 19.6mmol, 1.1eq) was added dropwise at -78 °C, the mixture was stirred for 2 h at this temperature, and 2-chloro-5-(chloromethyl)thiazole ( A13-1 , 3g, 34.7mmol, 1.0eq) dissolved in THF (10mL) was added dropwise to the system. Then the reaction continued at the temperature for 1.5 h. TLC testing indicated that a little raw material was unreacted.
  • reaction mixture was quenched with water, extracted with ethyl acetate (100mLx2).
  • the organic phases were combined and then washed with saturated sodium chloride aqueous solution.
  • Step 2 1-tert-butyl-4-ethyl 4-(2-chlorothiazol-5-yl)methyl)piperidine-1,4-dicarboxylate ( A13-2 , 4g, 10.3mmol) was dissolved in THF (100mL), and LDA(2M, 8.5mL, 16.6mmol) was added dropwise at -78 °C. The mixture was reacted for 1 h, and then the reaction was completed. Saturated amine chloride (100mL) was added to quench the reaction and the mixture was extracted with ethyl acetate (100mL ⁇ 3). The organic phases were combined and washed with saturated sodium chloride aqueous solution.
  • Step 3 2-chloro-4-oxo-4,6-dihydrospiro[cyclopenta[d]thiazole-5,4'-piperidine]-1'-carboxylic acid tert-butyl ester ( A13-3 , 710mg, 2.07mmol), Ti(OEt) 4 and (R)-2-methylpropane-2-sulfinamide (276mg, 2.28mmol) were added to a reaction flask, and the mixture was heated to 100°C under nitrogen protection for 5 h. The reaction was completed. The mixture was cooled to room temperature and diluted with ethyl acetate (50mL), saturated brine (15mL) was added, and white solid precipitated out. The mixture was filtered.
  • Step 4 Tert-butyl (E)-4-((tert-butylsulfinyl)imino)-2-chloro-4,6-dihydrospiro[cyclopenta[d]thiazole-5,4'-piperid ine]-1'-carboxylic acid tert-butyl ester ( A13-4 , 200mg, 0.47mmol) was dissolved in THF (10mL), and the mixture was cooled to -78 °C under nitrogen protection.
  • Step 5 Tert-butyl (S)-4-(((R)-tert-butylsulfinyl)amino)-2-chloro-4,6-dihydrospiro[cyclopenta[d]thiazole-5,4'-pip eridine]-1'-carboxylic acid tert-butyl ester (A13-5, 155mg, 0.35mmol) was dissolved in 4 M of dioxane hydrochloride solution (5mL, 20mmol), and the mixture was reacted at room temperature for 3 hours. The reaction was completed.
  • Step 1 2,4-dichloro-5-iodopyrimidine (1.1g, 4mmol) and 20mL of anhydrous ethanol were added to a dry 100mL flask. 80% hydrazine hydrate mixture (601mg, 12mmol) was slowly added thereto at 0 °C under the protection of nitrogen, and the mixture continued to be stirred for 1 hour. After the reaction was completed, the mixture was filtered and washed with anhydrous ethanol to obtain 2-chloro-4-hydrazino-5-iodopyrimidine ( B2-1 , 850mg, yield: 78.7%).
  • Step 2 2-chloro-4-hydrazino-5-iodopyrimidine (810mg, 3mmol) and trimethyl orthoformate (10mL) were added to a dried 100mL flask successively. Under the protection of nitrogen, the mixture was heated to 85 °C and stirred for 5 hours. After the reaction was completed, the obtained residue was poured into saturated NaCl solution (50mL), extracted with ethyl acetate (3 ⁇ 30mL), and washed with saturated brine. The organic layers were combined, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure.
  • Step 1 2,4-dichloro-5-iodopyrimidine (1.37g, 5mmol), 2,2-dimethoxyethylamine (8.4g, 10mmol) and anhydrous ethanol (50mL) were added to a dry 100mL flask successively. Then triethylamine (1.01g, 10mmol) was slowly added dropwsie to the reaction mixture under the protection of nitrogen at 0 °C, and then the mixture was stirred at room temperature for 10 hours. After the reaction was completed, the reaction mixture was concentrated in vacuum. 15mL of water was added to the obtained concentrate, and the mixture was extracted with dichloromethane (3 ⁇ 50mL) and washed with saturated brine.
  • Step 2 2-chloro-N-(2,2-dimethoxyethyl)-5-iodopyrimidine-4-amine ( B3-1 , 1.03g, 3mmol) and 10mL of concentrated sulfuric acid were added to a dried 100mL flask successively. Under the protection of nitrogen, the mixture was heated to 65 °C and stirred for 2 hours. After the reaction was completed, the reaction liquid was cooled to room temperature.
  • Step 3 8-iodoimidazo[1,2-c]pyrimidin-5-ol ( B3-2 , 522mg, 2mmol) and phosphorus oxychloride (8mL) were successively added to a dry 50mL single-necked flask. Under the protection of nitrogen, N,N-diisopropylethylamine (1mL) was slowly add dropwise, and then the mixture was heated to 120°C and stirred for 5 hours. After the reaction was completed, the reaction liquid was cooled to room temperature and concentrated in vacuum, then quenched with saturated sodium bicarbonate solution, extracted with ethyl acetate (3 ⁇ 40mL), dried over anhydrous sodium sulfate, filtered and concentrated.
  • the obtained residue was purified by silica gel chromatography (0-30% gradient of ethyl acetate: petroleum ether) to obtain 5-chloro-8-iodoimidazo[1,2-c]pyrimidine ( B3 , 360mg, yield: 55%) as a light yellow solid.
  • Step 1 3-chloro-4-iodopyridine-2-amine (2.5g, 9.82mmol, 1.0eq), XantPhos (341mg, 0.59mmol. 0.06eq), palladium acetate (110mg, 0.49mmol, 0.05eq), DIPEA (3.25ml, 19.6mmol, 2.0eq), methyl 3-mercaptopropionate (1.19ml, 10.8mmol, 1.1eq) and 1,4-dioxane (32.5mL) were added to a dry 100mL round-bottom three-necked flask successively. Under stirring, the atmosphere in the reaction system was replaced with nitrogen three times, and then the mixture was heated to 100 °C and reacted for 3 hours.
  • the reaction liquid was cooled to room temperature, diluted with ethyl acetate (50mL) and filtered under reduced pressure, and the filter cake was washed with ethyl acetate (25mL), The obtained filtrate was concentrated in vacuum, and the obtained residue was purified by silica gel chromatography (0-30% gradient of ethyl acetate: petroleum ether) to obtain methyl 3-((2-amino-3-chloropyridin-4-yl)thio)propionate ( C1-1 , 2.0g, yield: 78%) as a yellow solid.
  • Step 2 Compound C1-1 (2g, 8. 11mmol, 1.0eq) was dissolved in tetrahydrofuran (28mL) in a dry 100mL round-bottom three-necked flask, and sodium ethanol (2.9g, 8.51mmol, 1.05eq, 20% wt) was added dropwise to the reaction solution at room temperature under the protection of nitrogen, and then the mixture was stirred for one hour. After the reaction was complete, the mixture was diluted with dichloromethane (60mL) and treated with ultrasound for 5min, and then filtered under reduced pressure. The filter cake was dried in vacuum to obtain sodium 2-amino-3 chloropyridine-4-sulfide ( C1 , 1.4g, yield: 89%) as a yellow solid.
  • Step 1 2,3-dichloro-4-iodopyridine (1.0g, 3.65mmol, 1.0eq), methyl 3-mercaptopropionate (480mg, 4.02mmol, 1.1eq) and N,N-diisopropylethylamine (950mg, 7.3mmol) were dissolved in 1,4-dioxane (15mL), the atmosphere in the reaction system was replaced with argon three times, and palladium acetate (82mg, 0.37mmol, 0.1eq) and XantPhos (211mg, 0.37mmol, 0.1eq) were added under the protection of argon.
  • Step 2 Methyl 3-((2,3-dichloropyridin-4-yl)thio)propionate ( C2-1 , 100mg, 0.37mmol, 1.0eq) was dissolved in tetrahydrofuran (10mL) and ethanol (0.5mL). Sodium alcohol (27mg, 0.39mmol, 1.05eq) was added under the protection of nitrogen, the mixture was reacted at room temperature for 3 hours, the reaction was not completed, sodium alcohol (27mg, 0.39mmol, 1.05eq) was added, the mixture was reacted at room temperature for 3 hours, and then the reaction was completed.
  • Step 1 3-bromo-2-trifluoromethylpyridine (400mg, 1.77mmol, 1.0eq), methyl 3-mercaptopropionate (235mg, 1.95mmol, 1.1eq) and N,N-diisopropylethylamine (460mg, 3.54mmol, 2.0eq) was dissolved in 1,4-dioxane (15mL), the atmosphere in the reaction system was replaced with argon three times, and Pd 2 (dba) 3 (160mg, 0.18mmol, 0.1eq) and XantPhos (205mg, 0.36mmol, 0.2eq) were added under the protection of argon.
  • dba 160mg, 0.18mmol, 0.1eq
  • XantPhos 205mg, 0.36mmol, 0.2eq
  • Step 2 Methyl 3-((2-(trifluoromethyl)pyridin-3-yl)thio)propionate ( C3-1 , 300mg, 1.13mmol, 1.0eq) was dissolved in tetrahydrofuran (10mL) and ethanol (0.5mL). Sodium alcohol (84mg, 1.23mmol, 1.1eq) was added under the protection of nitrogen, the mixture was reacted at room temperature for 3 hours, the reaction was not completed, sodium alcohol (83mg, 1.23mmol, 1.1eq) was added, the mixture was reacted at room temperature for 3 hours, and then the reaction was completed. Tetrahydrofuran was removed under reduced pressure at room temperature.
  • Step 1 4-bromo-2-trifluoromethylpyridine (1.0g, 4.4mmol, 1.0eq), methyl 3-mercaptopropionate (760mg, 6.3mmol, 1.4eq) and N,N-diisopropylethylamine (2.17g, 16.8mmol, 3.8eq) were dissolved in 1,4-dioxane (25mL), the atmosphere in the reaction system was replaced with argon three times, and Pd 2 (dba) 3 (200mg, 0.22mmol, 0.05eq) and XantPhos (124mg, 0.22mmol, 0.05eq) were added under the protection of argon.
  • dba 200mg, 0.22mmol, 0.05eq
  • XantPhos 124mg, 0.22mmol, 0.05eq
  • Step 4 3-((2-(trifluoromethyl)pyridin-4-yl)thio)propionate (C4-1, 230mg, 0.85mmol, 1.0eq) was dissolved in tetrahydrofuran (10mL) and ethanol (0.5mL). Sodium alcohol (294mg, 0.87mmol, 1.02eq) was added under the protection of nitrogen, the mixture was reacted at room temperature for 1 hour, and the reaction was completed. Tetrahydrofuran was removed under reduced pressure at room temperature. Purified water was added and the mixture was freeze-dried to obtain a crude product of sodium 2-(trifluoromethyl)pyridine-4-sulfide (C4, 150mg) as a light yellow solid.
  • LCMS m/z 180.0 [M+H] +
  • Step 1 2-fluoro-4-iodopyridine (2.0g, 8.97mmol) was dissolved in THF (30mL). The atmosphere in the reaction system was replaced with nitrogen three times, then the temperature was reduced to -65 °C, and LDA(2.0 M in THF, 5.4ml, 10.80mmol) was added dropwise. The reaction solution gradually turned brown. After the addition of LDA was completed, the temperature was maintained for 1.5 hours. A solution of 1,3,2-dioxothiophene-2,2-dioxide (1.45g, 11.7mmol) in THF (30mL) was added dropwise. Then the mixture was warmed to room temperature naturally and stirred overnight.
  • reaction liquid was poured into saturated NaHCO 3 aqueous solution (50mL), extracted with ethyl acetate (3 ⁇ 30mL), washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and concentrated to dry, and then purified by silica gel chromatography to obtain 2-(2-fluoro-4-iodopyridin-3-yl)ethane-1-ol (C5-1, 2.07g, yield: 86.2%).
  • Step 3 Methyl 3-((3-chloro-2-(methylamino)pyridin-4-yl)thio)propionate (C6-2, 114mg, 0.44mmol) was dissolved in dioxane (6mL). Under the protection of argon, a solution of EtONa (20%(w/w), 150mg, 0.44mmol) in ethanol was added dropwise, and the mixture was stirred at room temperature for about 2.5 hours. LCMS indicated the reaction was completed. The reaction solution was directly used for the next reaction (yield: 100%). LCMS: m/z 174.8 [M+H-23] +
  • Step 1 Hydrazine hydrate (3032mg, 60.56mmol) was added to a solution of 2-fluoro-3-formyl-4-iodopyridine (1900mg, 7.57mmol) in isopropanol (30mL), and the mixture was stirred at 60°C for 3 h.
  • the reaction solution was concentrated under reduced pressure to remove part of the solvent, then poured into water, and filtered. The filter cake was washed with water to obtain 4-iodo-1H-pyrazolo[3,4-b]pyridine (C7-1, 1.8g, yield: 97%) as a light yellow solid.
  • Step 2 P-toluenesulfonic acid (28mg, 0.16mmol) and DHP (206mg, 2.45mmol) were added to a solution of 4-iodo-1H-pyrazolo[3,4-b]pyridine (C7-1, 400mg, 1.63mmol) in tetrahydrofuran (10mL) successively. The mixture was stirred at 60 °C for 16 h. The reaction solution was diluted with ethyl acetate (40mL), washed with saturated brine (2 ⁇ 40mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure.
  • Step 3 4-iodo-1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-B]pyridine (C7-2, 390mg, 1.18mmol), methyl 3-mercaptopropionate (157mg, 1.30mmol) and N,N-diisopropylethylamine (306mg, 2.37mmol) were dissolved in 1,4-dioxane (10mL), the atmosphere in the reaction system was replaced with argon three times, and Pd(OAc) 2 (27mg, 0.12mmol) and Xantphos (137mg, 0.24mmol) were added under the protection of argon.
  • Pd(OAc) 2 27mg, 0.12mmol
  • Xantphos 137mg, 0.24mmol
  • Step 4 Methyl 3-((1H-pyrazolo[3,4-b]pyridin-4-yl)thio)propionate (C7-3, 280mg, 0.87mmol) was dissolved in 1,4-dioxane (7mL) in a three-necked flask with a condenser tube. Under the protection of nitrogen, the mixture was cooled to 0°C, and sodium ethoxide ethanol solution (20%(w/w), 266mg, 0.78mmol) was added. The mixture was stirred for 2 h at room temperature.
  • Example 1 Synthesis of compound 1 (not encompassed by the present invention) (S)-1'-((2,3-dihydrofuran[2,3-b]pyridin-4-yl)thio)imidazolo[1,2-c]pyrimidin-5-yl)-5, 7-dihydrospirocyclopenta[b]pyridine-6,4'-piperidine]-5-amine
  • Step 1 Under the protection of nitrogen, 5-chloro-8-iodoimidazo[1,2-c]pyrimidine (B3, 80mg, 0.285mmol), ((R)-N-((S)-1,3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-methylpropane-2-sulfinamide (A4,105mg,0.342mmol), DIEA(55mg,0.428mmol) and CH 3 CN(5mL) were added to a dry 25mL single-necked flask successively, and then the mixture was stirred at 95°C for 3 hours. After the reaction was completed, the obtained reaction solution was filtered and concentrated under reduced pressure.
  • Step 2 Sodium 2,3-dihydrofuran[2,3-b]pyridin-4-thioformate (107mg, 0.61mmol) was added to dioxane (15mL) for dilution, and then (S)-N-((S)-1'-(8-iodoimidazolo[1,2-C]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopentane[B]pyrid in-6,4'-piperidin]-5-yl)-2-methylpropane-2-sulfinamide (1-1, 200mg, 0.36mmol), DIPEA (141mg, 1.09mmol), XantPhos (63mg, 0.11mmol) and Pd 2 (dba) 3 (50mg, 0.06mmol) were added.
  • Example 2 Synthesis of compound 2 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-1 ,3-dihydrospiro[indene-2,4'-piperidine]-1-amine
  • Step 1 Under the protection of nitrogen, 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine ( B2, 80mg, 0.285mmol), ((R)-N-((S)-1,3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-methylpropane-2-sulfinamide ( A1 , 105mg, 0.342mmol), DIEA (55mg, 0.428mmol) and CH 3 CN (5mL) were added to a dry 25mL single-necked flask successively, and then the mixture was stirred for 3 hours at 95 °C.
  • Step 2 Under the protection of nitrogen, (R)-N-((S)-1'-(8-iodo-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-1,3-dihydrospiro[indene-2,4'-piper idin]-1-yl)-2-methylpropane-2-sulfinamide ( 2-1, 100mg, 0.18mmol), sodium 2-amino-3-chloropyridine-4-mercaptan (49mg, 0.27mmol), Pd 2 (dba) 3 (16mg, 0.018mmol), Xantphos (21mg, 0.036mmol), DIPEA(58mg, 0.45mmol) and 1,4-dioxane solution (10mL) were added to a 5mL microwave reaction flask successively, and the mixture was microwaved to 100 °C and stirred for 3 hours under the protection of nitrogen.
  • Step 3 Under the protection of nitrogen, (R)-N-((R)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-1, 3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-methylpropane-2-sulfinamide (2-2, 60mg, 0.10mmol) and methanol (0.6mL) were added to a 50mL single-necked flask successively, hydrochloric acid 1,4-dioxane solution (0.06mL, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour.
  • hydrochloric acid 1,4-dioxane solution (0.06mL, 4M
  • Example 3 Synthesis of compound 3 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5 ,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine
  • Example 5 Synthesis of compound 5 (not encompassed by the present invention) (S)-1'-(8-((3-chloro-2-(methylamino)pyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl) -5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine
  • Step 1 sodium 2-methylamino-3-chloropyridin-4 sulfide (C6, 86mg) was dissolved in 10mL of dioxane solution, and (R)-N-((S)-1'-(8-iodoimidazolo[1,2-C]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridin-6,4'- piperidin]-5-yl)-2-methylpropane-2-sulfinamide (161mg, 0.29mmol), Pd 2 (dba) 3 (86mg, 0.094mmol), Xantphos (108mg, 0.19mmol) and DIPEA (400mg, 3.12mmol) were added.
  • Step 2 (R)-N-((s)-1'-(8-((3-chloro-2-(methylamino)pyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridin-6,4'-piperidin]-5-yl)-2-methylpropane-2-sulfinamide (135mg, 0.23mmol) was dissolved in methanol (20mL), HCl/dioxane (4 M, 1.2mL) was slowly added dropwise under the protection of nitrogen, and the mixture was reacted at room temperature for 40 minutes. LCMS testing indicated the reaction was completed.
  • Step 1 B3 (1.37g, 4.9mmol), A4 (1.35g, 4.9mmol) and DIPEA (4.86ml, 29.41mmol) were successively added to 3mL acetonitrile in a 25mL single-necked flask, and then the mixture was stirred for 2 hours at 80 °C. After the reaction was completed, the mixture was cooled to room temperature, then Boc 2 O (1.6g, 7.35mmol, 1.5eq) was added, and the mixture was heated to 50 °C and reacted until the reaction was completed. The reaction solution was concentrated under reduced pressure to obtain a residue.
  • Step 2 Under the protection of nitrogen, 6-1 (1.7g, 3.11mmol), sodium 2-amino-3-chloropyridine-4-mercaptan (596mg, 3.27mmol), Pd 2 (dba) 3 (285mg, 0.311mmol), Xantphos (360mg, 0.622mmol), DIPEA (804mg, 6.22mmol) and 1,4-dioxane solution (30mL) were added to a 5mL microwave reaction flask successively, and the mixture was microwaved to 100°C and stirred for 3 hours under the protection of nitrogen. After the reaction was completed, the mixture was cooled to room temperature, filtered and concentrated under reduced pressure to obtain a residue.
  • Step 3 Under the protection of nitrogen, TFA (5mL) was slowly added to a solution of (S)-tert-butyl (1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazolo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[ cyclopenta[b]pyridin-6,4'-piperidin]-5-yl)-carbonate ( 6-2, 1.2g, 2.07mmol) in dichloromethane (5mL) at 0 °C, the mixture was stirred at room temperature for 1 hour, and TLC and LCMS indicated the reaction was completed.
  • dichloromethane 5mL
  • reaction solution was concentrated under reduced pressure, then dichloromethane/methanol mixed solution was added for dissolution, and the pH of the mixture was adjusted to neutral with NaHCO 3 .
  • the mixture was purified by passing through silica gel column to obtain (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro [cyclopenta[b]pyridine-6,4'-piperidine]-5-amine (compound 6, 400mg, yield: 40.0%).
  • Example 7 Synthesis of compound 7 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5 ,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-7-amine
  • Example 8 Synthesis of compound 8 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihy drospiro[cyclopenta[c]pyridine-6,4'-piperidine]-7-amine
  • Example 9 Synthesis of compound 9 (not encompassed by the present invention) (S)-1'-(8-(2,3-dichloropyridin-4-yl)thio-[1,2,4]-triazolo[4,3-c]pyrimidin-5-yl)-5,7-dih ydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-7-amine
  • Step 1 (S)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-7-amine hydrochloride (A6, 200mg, 1.28mmol) was dissolved in acetonitrile (20mL), DIPEA (1.6mL, 9.68mmol) and 5-chloro-8-iodo-[1,2,4]-triazolo[4,3-c]pyrimidine (B2, 180mg, 0.64mmol) were added, the mixture was heated to 90 °C and refluxed for 3 hours under the protection of nitrogen, and the reaction was completed.
  • DIPEA 1.6mL, 9.68mmol
  • 5-chloro-8-iodo-[1,2,4]-triazolo[4,3-c]pyrimidine B2, 180mg, 0.64mmol
  • Step 2 (S)-1'-(8-iodo[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6, 4'-piperidine]-7-amine ( 21-1, 55mg, 0.12mmol) was dissolved in 1,4-dioxane (3mL), and DIPEA (35mg, 0.27mmol) and sodium 2,3-dichloropyridine-4-mercaptan (C2, 100mg, crude product) were added. XantPhos (30mg, 0.05mmol) and Pd 2 (dba) 3 (17mg, 0.02mmol) were added under the protection of nitrogen.
  • Example 10 Synthesis of compound 10 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihy drospiro[cyclopenta[b]pyridine-6,4'-piperidine]-7-amine
  • Example 11 Synthesis of compound 11 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5 ,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-7-amine
  • Example 12 Synthesis of compound 12 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-6 -fluoro-1,3-dihydrospiro[indene-2,4'-piperidine]-1-amine
  • Example 13 Synthesis of compound 13 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-6-fluoro -1,3-dihydrospiro[indene-2,4'-piperidine]-1-amine
  • Step 3 (S)-(1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5-fluoro-1,3-dih ydrospiro[indene-2,4'-piperidin]-3-yl)carboxylic acid tert-butyl ester ( 13-2, 60mg, 0.101mmol), dichloromethane (1mL) and trifluoroacetic acid (0.2mL) were added to a dry single-necked flask successively. The reaction solution was stirred at 20°C for 1 hour. The reaction liquid was concentrated under reduced pressure.
  • the pH of the obtained residual reaction liquid was adjusted to 8 with saturated sodium bicarbonate aqueous solution.
  • the mixture was extracted with DCM/MeOH (10:1) mixed solvent (10mL) for three times, and the combined organic phase was concentrated under reduced pressure.
  • the obtained residue was purified by HPLC preparation to obtained (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-6-fluoro-1,3-dih ydrospiro[indene-2,4'-piperidine]-1-amine ( 13, 32mg, formate, yield: 58.2%) as a white solid.
  • Example 14 Synthesis of compound 14 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihy drospiro[cyclopenta[c]pyridine-6,4'-piperidine]-5-amine
  • Step 1 (S)-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-5-amine hydrochloride ( A8 , 200mg, 0.83mmol) was dissolved in acetonitrile (20mL), DIPEA (1.07g, 8.3mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine( B3 , 208mg, 0.75mmol) were added, the mixture was heated to 90 °C and refluxed for 5 hours under nitrogen protection, and the reaction was completed. The reaction liquid was cooled to room temperature, poured into saturated sodium bicarbonate aqueous solution, and extracted twice with dichloromethane (50mL).
  • Step 9 1'-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidi ne]-4-amine ( 14-1 , 50mg, 0.11mmol) was dissolved in 1,4-dioxane (10mL), DIPEA (36mg, 0.28mmol) and sodium 2-amino-3-chloropyridine-4-mercaptan ( C1 , 31mg, 0.17mmol) were added. XantPhos (13mg, 0.02mmol) and Pd 2 (dba) 3 (10mg, 0.01mmol) were added under the protection of nitrogen.
  • Example 15 Synthesis of compound 15 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5 ,7-dihydrospiro[cyclopenta[c]pyridine-6,4'-piperidine]-5-amine
  • Example 16 Synthesis of compound 16 (not encompassed by the present invention) (S)-1-amino-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimid in-5-yl)-1,3-dihydrospiro[indene-2,4'-piperidin]-4-ol
  • Step 1 (S)-1-amino-1,3-dihydrospiro[indene-2,4'-piperidin]-4-ol hydrochloride ( A9 , 200mg, 0.79mmol) was dissolved in acetonitrile (40mL), DIPEA (1.07g, 8.3mmol) and 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine (B2, 208mg, 0.75mmol) were added, the mixture was reacted at room temperature under nitrogen protection for 18 hours, and the reaction was completed. The reaction liquid was cooled to room temperature, poured into saturated sodium bicarbonate aqueous solution, and extract with dichloromethane (100mL) twice.
  • Step 2 (S)-1-amino-1'-(8-iodo-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-1,3-dihydrospiro[indene-2,4'-pip eridin]-4-ol ( 16-1, 200mg, 0.43mmol) was dissolved in 1,4-dioxane (30mL), and DIPEA (111mg, 0.86mmol) and sodium 2-amino-3-chloropyridin-4-mercaptan ( C1 , 118mg, 0.65mmol) were added.
  • DIPEA 111mg, 0.86mmol
  • sodium 2-amino-3-chloropyridin-4-mercaptan C1 , 118mg, 0.65mmol
  • Example 17 Synthesis of compound 17 (not encompassed by the present invention) Compound (S)-1-amino-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-1,3-dihydrospiro[indene-2,4'-piperidine]-4-nitrile
  • Step 1 (S)-1-(((R)-tert-butylsulfinyl)amino)-4-cyano-1,3-dihydrospiro[indene-2,4'-piperidine] ( A10 , 490mg, 1.48mmol) was dissolved in acetonitrile (20mL), DIPEA(1.91g, 14.8mmol) and 5-chloro-8-iodo -[1,2,4]triazolo[4,3-c]pyrimidine ( B2, 415mg, 1.48mmol) were added, the mixture was reacted at 85°C under nitrogen protection for 3 hours, and the reaction was completed.
  • Step 2 (R)-N-((S)-1'-(8-iodo-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4-cyano-1,3-dihydrospiro[indene-2,4'- piperidin]-1-yl)-2-tert-butylsulfonamide ( 17-1, 360mg, 0.63mmol) was dissolved in 1,4-dioxane (10mL), and DIPEA(202mg, 1.56mmol) and sodium 2-amino-3-chloropyridin-4-mercaptan ( C1 , 171mg, 0.94mmol ) were added.
  • Step 3 (R)-N-((S)-1'-((8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4 -cyano-1,3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-tert-butylsulfonamide ( 17-2, 130mg, 0.21mmol) and methanol (6mL) were successively added to a 50mL single-necked flask under the protection of nitrogen, and hydrochloric acid 1,4-dioxane solution (2ml, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour.
  • Example 18 Synthesis of compound 18 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihy drospiro[cyclopenta[b]pyrazine-6,4'-piperidine]-5-amine
  • Step 1 (S)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine hydrochloride ( A11 , 1.50g, 4.95mmol) was dissolved in 150mL of acetonitrile, DIPEA (5.68g, 44.0mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine ( B3, 1.28g, 4.59mmol) were added, the mixture was heated to 95°C for and refluxed for 3 hours under nitrogen protection, and the reaction was completed. The mixture was cooled to room temperature and filtered.
  • Step 2 (S)-1'-(8-iodoimidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyrazine-6,4'-pipe ridine]-5-amine ( 18-1, 2.05g, 4.59mmol) was dissolved in 120mL of 1,4-dioxane in a 350mL glass seal tube, DIPEA (1.78g, 13.8mmol), sodium 2-amino-3-chloropyridine-4-mercaptan ( C1 , 1.26g, 6.9mmol), XantPhos (797mg, 1.38mmol) and Pd 2 (dba) 3 (630mg, 0.69mmol) were added successively, the reaction mixture was sparged with argon for 30 seconds, and was heated to 100 °C under argon and maintained for 3 hours until the reaction was completed.
  • Example 19 Synthesis of compound 19 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4 -methoxy-1,3-dihydrospiro [indene-2,4'-piperidine]-1-amine
  • Step 7 (S)-1-(((R)-tert-butylsulfinyl)amino)-4-methoxy-1,3-dihydrospiro[indene-2,4'-piperidine] ( A12, 520mg, 1.55mmol) was dissolved in acetonitrile (20mL), DIPEA (2.0g, 15.5mmol) and 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine ( B2 , 435mg, 1.55mmol) were added, the mixture was reacted at 85 °C for 3 hours under the protection of nitrogen, and the reaction was completed.
  • DIPEA 2.0g, 15.5mmol
  • 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine B2 , 435mg, 1.55mmol
  • Step 8 (R)-N-((S)-1'-(8-iodo-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4-methoxy-1,3-dihydrospiro[inden e-2,4'-piperidin]-1-yl)-2-tert-butylsulfonamide ( 19-1, 420mg, 0.72mmol) was dissolved in 1,4-dioxane (10mL), and DIPEA (234mg, 1.81mmol) and sodium 2-amino-3-chloropyridine-4-mercaptan ( C1 , 198mg, 1.09mmol) were added.
  • reaction solution was concentrated and then purified by silica gel chromatography to obtain (R)-N-((S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4-methoxy-1,3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-tert-butylsulfonamide ( 19-2, 3 10mg, yield: 69.9%) as a white solid.
  • Step 9 (R)-N-((S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-4-methoxy-1,3-dihydrospiro[indene-2,4'-piperidin]-1-yl)-2-tert-butylsulfonamide ( 19-2, 310mg, 0.51mmol) and methanol (6mL) were added to a 50mL single-necked flask under the protection of nitrogen successively, hydrochloric acid 1,4-dioxane solution (2mL, 4M) was added dropwise at room temperature, and the mixture was stirred at room temperature for 1 hour.
  • Example 20 Synthesis of compound 20 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5 ,7-dihydrospiro[cyclopenta[b]pyrazine-6,4'-piperidine]-5-amine
  • Step 1 (S)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine hydrochloride ( A11 , 89mg, 0.29mmol) was dissolved in 20mL of acetonitrile, DIPEA (212mg, 1.64mmol) and 5-chloro-8-iodo-[1,2,4]triazolo[4,3-c]pyrimidine ( B2, 77mg, 0.275mmol) were added, the mixture was heated to 95°C and refluxed for 3 hours under the protection nitrogen, and the reaction was completed. The mixture was cooled to room temperature and filtered.
  • Step 2 (S)-1'-(8-iodo-[1,2,4]triazolo[4,3-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyrazine-6 ,4'-piperidine]-5-amine (20-1, 65mg, 0.145mmol) was dissolved in 5mL of 1,4-dioxanein a 5mL microwave tube, and DIPEA (56mg, 0.434mmol), 2-amino-3-chloropyridine-4 sodium sulfide ( C1 , 40mg, 0.219mmol), XantPhos (34mg, 0.059mmol) and Pd 2 (dba) 3 (27mg, 0.029mmol) were added successively, the reaction mixture was sparged with argon for 30 seconds, and was heated to 100°C under the atmosphere of argon and maintained for 3 hours until the reaction was completed.
  • DIPEA 56mg, 0.434m
  • Example 21 Synthesis of compound 21 (not encompassed by the present invention) (S)-1'-(8-((2-(trifluoromethyl)pyridin-3-yl)thio)-[1,2,4]-triazolo[4,3-c]pyrimidin-5-yl )-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-7-amine
  • Example 22 Synthesis of compound 22 (not encompassed by the present invention) (S)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,5-c]pyrimidin-5-yl)-2-chlor o-4,6-dihydrospiro[cyclopenta[d]thiazole-5,4'-piperidine]-4-amine
  • Step 1 (S)-2-chloro-4,6-dihydrospiro[cyclopenta[d]thiazole-5,4'-piperidine]-4-amine hydrochloride (A13, 155mg, 0.35mmol) was dissolved in acetonitrile (10mL), and DIPEA (450mg, 3.5mmol) and 5-chloro-8-iodoimidazo[1,2-c]pyrimidine ( B3, 108mg, 0.39mmol) were added, the mixture was heated to 100°C and refluxed for 5 hours under the protection of nitrogen, and the reaction was completed.
  • DIPEA 450mg, 3.5mmol
  • 5-chloro-8-iodoimidazo[1,2-c]pyrimidine B3, 108mg, 0.39mmol
  • Step 2 (S)-2-chloro-1'-(8-iodoimidazo[1,2-c]pyrimidine-5-yl)-4,6-dihydrospiro[cyclopenta[d]thiazol e-5,4'-piperidine]-4-amine (22-1, 50mg, 0.1mmol) was dissolved in 1,4-dioxane (5mL), and DIPEA (30mg, 0.25mmol) and sodium 2-amino-3-chloropyridine-4-mercaptan ( C1 , 30mg, 0.15mmol) were added. XantPhos (13mg, 0.02mmol) and Pd 2 (dba) 3 (20mg, 0.02mmol) were added under the protection of nitrogen.
  • Step 1 Dioxane (20mL) was added to sodium 1-(tetrahydro-2H-pyran-2-yl)-1H-pyrazolo[3,4-b]pyridine-4-mercaptan (C7, 224mg, 0.8mmol) in a reaction flask, and then (S)-N-((S)-1'-(8-iodoimidazolo[1,2-C]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopentane[B]pyrid ine-6,4'-piperidin]-5-yl)-2-methylpropane-2-sulfinamide (1-1, 287mg, 0.52mmol), DIPEA (304mg, 2.35mmol), XantPhos (91mg, 0.16mmol) and Pd 2 (dba) 3 (72mg, 0.08mmol) were added.
  • the reaction solution was diluted with ethyl acetate and filtered. The filter residue was eluted with ethyl acetate.
  • Example 25 of WO2018136265 the closest control compound to the disclosure is Example 25 of WO2018136265 . According to the synthetic route and operation steps of Example 25 of WO2018136265 , the control compound aa was obtained.
  • the biological function of the compound of the disclosure has been proved by the tests on enzyme activity and at the celluar level.
  • the compound of the disclosure can achieve strong inhibition activity (IC50 can reach InM).
  • the compound of the disclosure also showed a very good activity on inhibiting the proliferation of cancer cells, and the inhibitory activity on the proliferation of MV4-11 cell line can reach InM.
  • SHP099 6-(4-amino-4-methylpiperidine-1-yl)-3-(2,3-dichlorophenyl)pyrazine-2-amine
  • the compound of the disclosure showed superior activity both at the enzymatic level and at the cellular level.
  • Test example 1 Test method of SHP2 enzyme activity
  • Test method of SHP2 enzyme activity Compound powder was dissolved in DMSO to make mother liquor. During the experiment, the compound stock solution was diluted with DMSO by 3-fold gradient, and 10 different test concentrations were set for each compound. 1 ⁇ L of the compounds at each concentration point was put into the well of the detection plate (Corning, Costa 3915), and each concentration was tested in two replicates. 6,8-difluoro-4-methyl-7-hydroxycoumarin phosphate (DiFMUP) was used as the substrate, and it can be hydrolyzed under the catalysis of SHP2 E72A to produce 6,8-difluoro-4-methyl-7-hydroxycoumarin (DiFMU). To determine the enzyme activity of SHP2, the fluorescence value at 455 nm was detected by PE Enspire multifunctional reader using an excitation wavelength of 358 nm.
  • DiFMUP 6,8-difluoro-4-methyl-7-hydroxycoumarin phosphate
  • the SHP2 buffer for the reaction consisted of 60mmol/L Hepes, PH7.2, 75mmol/L NaCl, 75mmol/L KCl, 1mmol/L EDTA, 5mmol/L DTT.
  • the screening system consisted of SHP2 buffer, enzyme SHP2 E76A protein, substrate DiFMUP and test compounds.
  • Test method of IC50 In a 96-well screening plate, 50ng SHP2 E76A protein was reacted with the test compound in SHP2 buffer for 20 min, and then incubated with 10uM DiFMUP at room temperature for 20 min. The light intensity at 455nm was read by PE Enspire multifunction reader with an excitation light of 358 nm. The inhibition rate of the sample on the enzyme activity was calculated as the ratio of the measured fluorescence value of the compound treatment group to that of the DMSO control well. IC50 values of the compounds were calculated using Graphpad's Prism software by nonlinearly fitting the inhibition rate to the concentration of the inhibitor.
  • Test example 2 MV4-11 cell proliferation inhibition experiment
  • the number of living cells in the culture was determined by quantitative determination of intracellular ATP using CellTiter-Glo ® Luminescent Cell Viability Assay kit.
  • MV4-11 cells were inoculated into 96-well plates with a density of 2500 cells per well and a volume of 100 ⁇ L per well. The plates were placed in 37°C, 5% carbon dioxide incubator overnight.
  • cells were treated with compounds.
  • the compounds to be tested were diluted 3 times, and 8 concentration gradients were set up.
  • Each well was added with a certain volume of DMSO or the compound to be tested, and each concentration was tested in 2 replicates, and the final concentration of DMSO was 0.5%.
  • the plates were placed in 37°C, 5% carbon dioxide incubator for 72h.
  • Pharmacokinetics of the compound of the disclosure was determined. The following method was used to determine the pharmacokinetic parameters of the compound of the disclosure.
  • mice Healthy male adult mice were used in the study. Each group was intragastrically administered with a single dose of 5-100mg/kg. Fasting lasted from 10 hours before administration to 4 hours after administration. Blood samples were collected at different time points after administration and the compound content in plasma was determined by LC-MS/MS. The relationship between plasma concentration and time was analyzed by professional software (winnonlin), and the pharmacokinetic parameters of the compounds were calculated. According to Table 3, the compounds of the disclosure have excellent pharmacokinetic properties.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Claims (11)

  1. Composé ayant une structure :
    Figure imgb0064
    ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci.
  2. Composition pharmaceutique comprenant :
    (i) une quantité efficace du composé selon la revendication 1 ou d'un sel pharmaceutiquement acceptable, d'un énantiomère, d'un diastéréoisomère, d'un tautomère, d'un solvate ou d'un dérivé substitué par un isotope de celui-ci ; et
    (ii) un véhicule pharmaceutiquement acceptable.
  3. Composé selon la revendication 1, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation dans la prévention ou le traitement d'une maladie ou pathologie, dans lequel la maladie ou pathologie est un cancer.
  4. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 3, dans lequel :
    (a) la maladie ou pathologie est liée à une activité SHP2 anormale ;
    (b) la maladie ou pathologie est une maladie ou pathologie médiée par SHP2 ; ou
    (c) l'activité SHP2 est inhibée.
  5. Utilisation du composé selon la revendication 1 ou d'un sel pharmaceutiquement acceptable, d'un énantiomère, d'un diastéréoisomère, d'un tautomère, d'un solvate ou d'un dérivé substitué par un isotope de celui-ci dans :
    (d) l'inhibition de l'activité SHP2 de manière non thérapeutique in vitro ; ou
    (e) l'inhibition de la prolifération des cellules tumorales de manière non thérapeutique in vitro.
  6. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 3, dans lequel la maladie ou pathologie est choisie dans le groupe constitué par le syndrome de Noonan, le syndrome Léopard, une leucémie myélomonocytaire juvénile, un neuroblastome, un mélanome, une leucémie myéloïde aiguë, un cancer du sein, un cancer de l'œsophage, un cancer du poumon, un cancer du côlon, un cancer de la tête, un neuroblastome, un carcinome épidermoïde de la tête et du cou, un cancer gastrique, un lymphome anaplasique à grandes cellules et un glioblastome.
  7. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 4, dans lequel la maladie ou pathologie médiée par SHP2 est un cancer choisi dans le groupe constitué par une LMMJ, une LMA, un SMD, une LLA-B, un neuroblastome, un cancer de l'œsophage, un cancer du sein, un cancer du poumon, un cancer du côlon, un cancer de l'estomac, un cancer de la tête et du cou.
  8. Composé selon la revendication 1, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation simultanément, séquentiellement ou séparément en association avec une ou plusieurs substances choisies parmi un agent chimiothérapeutique ou des agents anticancéreux.
  9. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 8, dans lequel l'agent chimiothérapeutique est un inhibiteur mitotique.
  10. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 9, dans lequel l'inhibiteur mitotique est choisi dans le groupe constitué par les taxanes, les alcaloïdes de la pervenche, le paclitaxel, le docétaxel, la vincristine, la vinblastine, la vinorelbine et la vinflunine.
  11. Composé, ou un sel pharmaceutiquement acceptable, un énantiomère, un diastéréoisomère, un tautomère, un solvate ou un dérivé substitué par un isotope de celui-ci pour une utilisation selon la revendication 8, dans lequel l'agent anticancéreux est choisi dans le groupe constitué par le cisplatine, le 5-fluorouracil, le flutamide et la gemcitabine.
EP19881313.1A 2018-11-06 2019-11-05 (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine en tant qu'inhibiteur de shp2 pour le traitement du cancer Active EP3712151B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP22159530.9A EP4074713A1 (fr) 2018-11-06 2019-11-05 Dérivés de 1'-imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-pipéridine]-5-amine et composés similaires en tant qu'inhibiteurs de shp2 pour le traitement du cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201811314910 2018-11-06
PCT/CN2019/115754 WO2020094018A1 (fr) 2018-11-06 2019-11-05 Composé spiro cyclique aromatique et utilisation associée

Related Child Applications (1)

Application Number Title Priority Date Filing Date
EP22159530.9A Division EP4074713A1 (fr) 2018-11-06 2019-11-05 Dérivés de 1'-imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-pipéridine]-5-amine et composés similaires en tant qu'inhibiteurs de shp2 pour le traitement du cancer

Publications (3)

Publication Number Publication Date
EP3712151A1 EP3712151A1 (fr) 2020-09-23
EP3712151A4 EP3712151A4 (fr) 2020-12-30
EP3712151B1 true EP3712151B1 (fr) 2022-03-02

Family

ID=70516741

Family Applications (2)

Application Number Title Priority Date Filing Date
EP19881313.1A Active EP3712151B1 (fr) 2018-11-06 2019-11-05 (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine en tant qu'inhibiteur de shp2 pour le traitement du cancer
EP22159530.9A Pending EP4074713A1 (fr) 2018-11-06 2019-11-05 Dérivés de 1'-imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-pipéridine]-5-amine et composés similaires en tant qu'inhibiteurs de shp2 pour le traitement du cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP22159530.9A Pending EP4074713A1 (fr) 2018-11-06 2019-11-05 Dérivés de 1'-imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-pipéridine]-5-amine et composés similaires en tant qu'inhibiteurs de shp2 pour le traitement du cancer

Country Status (11)

Country Link
US (3) US10844079B2 (fr)
EP (2) EP3712151B1 (fr)
JP (1) JP2022507114A (fr)
KR (1) KR20210088639A (fr)
CN (4) CN111138412B (fr)
AU (1) AU2019374194A1 (fr)
BR (1) BR112021008714A2 (fr)
CA (1) CA3118925A1 (fr)
ES (1) ES2914852T3 (fr)
SG (1) SG11202104752RA (fr)
WO (1) WO2020094018A1 (fr)

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3515916B1 (fr) 2016-09-22 2023-06-07 Relay Therapeutics, Inc. Inhibiteurs de phosphatase shp2 et leurs procédés d'utilisation
TW201819386A (zh) 2016-10-24 2018-06-01 美商傳達治療有限公司 Shp2磷酸酶抑制劑及其使用方法
EP3630770A1 (fr) 2017-05-26 2020-04-08 Relay Therapeutics, Inc. Dérivés de pyrazolo[3,4-b]pyrazine en tant qu'inhibiteurs de la phosphatase shp2
US11701354B2 (en) 2017-09-29 2023-07-18 D. E. Shaw Research, Llc Pyrazolo[3,4-b]pyrazine derivatives as SHP2 phosphatase inhibitors
AU2019222026B2 (en) * 2018-02-13 2022-05-12 Shanghai Blueray Biopharma Co., Ltd. Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
CN115448923B (zh) * 2018-02-13 2024-03-22 上海青煜医药科技有限公司 嘧啶并环化合物及其制备方法和应用
TW202003471A (zh) 2018-03-21 2020-01-16 美商傳達治療有限公司 Shp2磷酸酶抑制劑及其使用方法
WO2020072656A1 (fr) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Dérivés d'imidozopyrimidine
CN111138412B (zh) 2018-11-06 2023-09-15 上海奕拓医药科技有限责任公司 一种螺芳环化合物及其应用
JP2022506887A (ja) * 2018-11-07 2022-01-17 シャンハイ リンジーン バイオファーマ カンパニー リミテッド 窒素含有縮合複素環系shp2阻害剤化合物、製造方法及び使用
BR112021009880A2 (pt) * 2018-11-30 2021-08-17 Tuojie Biotech (Shanghai) Co., Ltd. pirimidina e derivado de heterociclo de nitrogênio de cinco membros, método de preparação para os mesmos e usos médicos dos mesmos
CN111704611B (zh) * 2019-07-25 2022-01-14 上海凌达生物医药有限公司 一类芳基螺环类shp2抑制剂化合物、制备方法和用途
EP3772513A1 (fr) 2019-08-09 2021-02-10 C.N.C.C.S. S.c.a.r.l. Collezione Nazionale Dei Composti Chimici e Centro Screening Inhibiteurs d'shp2
EP4034539A1 (fr) 2019-09-24 2022-08-03 Relay Therapeutics, Inc. Inhibiteurs de phosphatase shp2, procédés de production et d'utilisation associés
CN112778276A (zh) * 2019-11-08 2021-05-11 南京圣和药业股份有限公司 作为shp2抑制剂的化合物及其应用
CN114846005B (zh) * 2020-01-21 2024-04-02 贝达药业股份有限公司 Shp2抑制剂及其应用
WO2021148010A1 (fr) * 2020-01-22 2021-07-29 南京明德新药研发有限公司 Composé à cycle pyrazolo hétéroaryl et son application
CN113754683A (zh) * 2020-06-05 2021-12-07 上海奕拓医药科技有限责任公司 同位素取代的螺芳环化合物及其应用
JP2023528990A (ja) * 2020-06-11 2023-07-06 貝達薬業股▲ふん▼有限公司 Shp2阻害剤、ならびにその組成物および使用
JP2023532217A (ja) 2020-06-18 2023-07-27 上海奕拓醫藥科技有限責任公司 Shp2阻害剤の結晶形、その組成物、その製造方法及び応用
IL299131A (en) 2020-06-18 2023-02-01 Revolution Medicines Inc Methods for delaying, preventing and treating acquired resistance to RAS inhibitors
EP4208261A1 (fr) 2020-09-03 2023-07-12 Revolution Medicines, Inc. Utilisation d'inhibiteurs de sos1 pour traiter des malignités à mutations de shp2
EP4214209A1 (fr) 2020-09-15 2023-07-26 Revolution Medicines, Inc. Dérivés d'indole servant d'inhibiteurs dans le traitement du cancer
CN113372344B (zh) * 2020-12-24 2022-11-01 上海药坦药物研究开发有限公司 一种氯代六元含氮杂环并咪唑类化合物的合成方法
WO2022156765A1 (fr) * 2021-01-22 2022-07-28 南京明德新药研发有限公司 Composé tricyclique lié à la pyrazolopyrazine et son application
PE20240634A1 (es) * 2021-02-05 2024-03-26 Hutchmed Ltd Compuestos triciclicos y usos de los mismos
CN113480536B (zh) * 2021-04-21 2024-02-23 中国人民解放军海军军医大学 螺环哌啶酮类衍生物
EP4334321A1 (fr) 2021-05-05 2024-03-13 Revolution Medicines, Inc. Inhibiteurs de ras
WO2022235866A1 (fr) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Inhibiteurs de ras covalents et leurs utilisations
CN117616031A (zh) 2021-05-05 2024-02-27 锐新医药公司 用于治疗癌症的ras抑制剂
CN115340545A (zh) * 2021-05-14 2022-11-15 浙江海正药业股份有限公司 双环杂芳基类衍生物及其制备方法和用途
WO2022241975A1 (fr) * 2021-05-20 2022-11-24 Etern Biopharma (Shanghai) Co., Ltd. Procédés de traitement de cancers associés à une mutation d'egfr
WO2022259157A1 (fr) 2021-06-09 2022-12-15 Novartis Ag Combinaison pharmaceutique triple comprenant du dabrafenib, du trametinib et un inhibiteur de shp2
TW202317100A (zh) 2021-06-23 2023-05-01 瑞士商諾華公司 包含kras g12c抑制劑的藥物組合及其用於治療癌症之用途
KR20240055778A (ko) 2021-09-01 2024-04-29 노파르티스 아게 Tead 억제제를 포함하는 제약 조합물 및 암의 치료를 위한 이의 용도
AR127308A1 (es) 2021-10-08 2024-01-10 Revolution Medicines Inc Inhibidores ras
WO2023109761A1 (fr) * 2021-12-15 2023-06-22 贝达药业股份有限公司 Cristal de composé pyrazolopyrimidinone et sel de celui-ci
WO2023172940A1 (fr) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Méthodes de traitement du cancer du poumon réfractaire immunitaire
WO2023230968A1 (fr) * 2022-06-01 2023-12-07 上海凌达生物医药有限公司 Inhibiteur de shp2, forme cristalline de celui-ci, procédé de préparation correspondant et utilisation associée
WO2023240263A1 (fr) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Inhibiteurs de ras macrocycliques
CN115267037B (zh) * 2022-07-25 2024-02-23 宁波熙宁检测技术有限公司 一种测定血浆中shp099浓度的方法

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE602005026546D1 (de) * 2004-09-29 2011-04-07 Mitsubishi Tanabe Pharma Corp Der tau-proteinkinase-1
ES2699351T3 (es) 2014-01-17 2019-02-08 Novartis Ag Derivados de 1-piridazin/triazin-3-il-piper(-azina)/idina/pirolidina y composiciones de las mismas para inhibir la actividad de SHP2
JO3517B1 (ar) * 2014-01-17 2020-07-05 Novartis Ag ان-ازاسبيرو الكان حلقي كبديل مركبات اريل-ان مغايرة وتركيبات لتثبيط نشاط shp2
EP3310771B1 (fr) * 2015-06-19 2020-07-22 Novartis AG Composés et compositions pour l'inhibition de l'activité de shp2
ES2741746T3 (es) * 2015-06-19 2020-02-12 Novartis Ag Compuestos y composiciones para inhibir la actividad de SHP2
CN112625028A (zh) 2015-06-19 2021-04-09 诺华股份有限公司 用于抑制shp2活性的化合物和组合物
CN107286150B (zh) 2016-04-11 2020-07-07 中国科学院上海有机化学研究所 N-杂环类化合物、其中间体、制备方法、药物组合物和应用
CA3026784A1 (fr) * 2016-06-07 2017-12-14 Jacobio Pharmaceuticals Co., Ltd. Derives de pyrazine heterocycliques utiles en tant qu'inhibiteurs de shp2
MX2018015625A (es) 2016-06-14 2019-03-06 Novartis Ag Compuestos y composiciones para inhibir la actividad de shp2.
EP3515916B1 (fr) 2016-09-22 2023-06-07 Relay Therapeutics, Inc. Inhibiteurs de phosphatase shp2 et leurs procédés d'utilisation
EP3571189B1 (fr) * 2017-01-23 2023-03-29 Revolution Medicines, Inc. Composés de pyridine utilisés en tant qu'inhibiteurs allostériques de shp2
CN108341791B (zh) 2017-01-23 2020-09-29 中国科学院上海药物研究所 苯并噻二唑类化合物、其制备方法及用途
JP7240319B2 (ja) * 2017-01-23 2023-03-15 レヴォリューション・メディスンズ,インコーポレイテッド アロステリックshp2阻害剤としての二環式化合物
CN108570048B (zh) * 2017-03-10 2021-06-08 广东东阳光药业有限公司 取代的杂芳基化合物及其组合物和用途
EP3601239A4 (fr) * 2017-03-23 2020-05-13 Jacobio Pharmaceuticals Co., Ltd. Nouveaux dérivés hétérocycliques utiles en tant qu'inhibiteurs de shp2
WO2019075265A1 (fr) 2017-10-12 2019-04-18 Revolution Medicines, Inc. Pyridine, pyrazine et composés de triazine utilisés en tant qu'inhibiteurs allostériques de shp2
MX2020006273A (es) 2017-12-15 2020-09-14 Revolution Medicines Inc Compuestos policiclicos como inhibidores alostericos de shp2.
AU2019222026B2 (en) 2018-02-13 2022-05-12 Shanghai Blueray Biopharma Co., Ltd. Pyrimidine-fused cyclic compound, preparation method therefor and application thereof
TW202003471A (zh) * 2018-03-21 2020-01-16 美商傳達治療有限公司 Shp2磷酸酶抑制劑及其使用方法
CN112203689A (zh) 2018-04-10 2021-01-08 锐新医药公司 治疗癌症的shp2抑制剂组合物和方法
CN110143949A (zh) * 2018-05-09 2019-08-20 北京加科思新药研发有限公司 可用作shp2抑制剂的新型杂环衍生物
WO2020063760A1 (fr) 2018-09-26 2020-04-02 Jacobio Pharmaceuticals Co., Ltd. Nouveaux dérivés hétérocycliques utiles en tant qu'inhibiteurs de shp2
WO2020072656A1 (fr) 2018-10-03 2020-04-09 Gilead Sciences, Inc. Dérivés d'imidozopyrimidine
CN111138412B (zh) 2018-11-06 2023-09-15 上海奕拓医药科技有限责任公司 一种螺芳环化合物及其应用
JP2022506887A (ja) * 2018-11-07 2022-01-17 シャンハイ リンジーン バイオファーマ カンパニー リミテッド 窒素含有縮合複素環系shp2阻害剤化合物、製造方法及び使用
BR112021009880A2 (pt) 2018-11-30 2021-08-17 Tuojie Biotech (Shanghai) Co., Ltd. pirimidina e derivado de heterociclo de nitrogênio de cinco membros, método de preparação para os mesmos e usos médicos dos mesmos
CN111704611B (zh) * 2019-07-25 2022-01-14 上海凌达生物医药有限公司 一类芳基螺环类shp2抑制剂化合物、制备方法和用途

Also Published As

Publication number Publication date
BR112021008714A2 (pt) 2021-08-10
CA3118925A1 (fr) 2020-05-14
CN111138412A (zh) 2020-05-12
US10844079B2 (en) 2020-11-24
US20240116949A1 (en) 2024-04-11
US20210238196A1 (en) 2021-08-05
CN111592525A (zh) 2020-08-28
CN111138412B (zh) 2023-09-15
SG11202104752RA (en) 2021-06-29
CN111592525B (zh) 2020-12-08
ES2914852T3 (es) 2022-06-17
US11685748B2 (en) 2023-06-27
EP3712151A1 (fr) 2020-09-23
WO2020094018A1 (fr) 2020-05-14
EP3712151A4 (fr) 2020-12-30
JP2022507114A (ja) 2022-01-18
EP4074713A1 (fr) 2022-10-19
AU2019374194A1 (en) 2021-05-27
CN111566104B (zh) 2024-05-17
CN111566104A (zh) 2020-08-21
CN117143079A (zh) 2023-12-01
US20200317695A1 (en) 2020-10-08
KR20210088639A (ko) 2021-07-14

Similar Documents

Publication Publication Date Title
EP3712151B1 (fr) (s)-1'-(8-((2-amino-3-chloropyridin-4-yl)thio)imidazo[1,2-c]pyrimidin-5-yl)-5,7-dihydrospiro[cyclopenta[b]pyridine-6,4'-piperidine]-5-amine en tant qu'inhibiteur de shp2 pour le traitement du cancer
CN111153901B (zh) 一类含氮稠杂环类shp2抑制剂化合物、制备方法和用途
CN110156786B (zh) 嘧啶并环化合物及其制备方法和应用
CN112300160A (zh) 一种螺芳环化合物、其制备及应用
CN112142735B (zh) 一类稠和氰基吡啶类化合物、制备方法和用途
WO2019158019A1 (fr) Composé cyclique fusionné à une pyrimidine, son procédé de préparation et son application
WO2021244659A1 (fr) Composé cyclique spiro aromatique substitué par un isotope et son application
CN110950876B (zh) 一类呋喃并内酰胺类化合物、制备方法和用途
CN113061132A (zh) 一类稠环内酰胺类化合物、制备方法和用途
CN112094269B (zh) 一类饱和六元环并杂环类化合物、制备方法和用途
CN110655520A (zh) 嘧啶并环化合物及其制备方法和应用
CN109790160B (zh) 吡啶并五元芳香环类化合物、其制备方法及用途
CN112300173B (zh) 一类含氮多环类化合物、制备方法和用途
WO2021083383A1 (fr) Composé cyclique fusionné contenant de l'azote en tant que régulateur de sting, son procédé de préparation et son utilisation
CN109761986B (zh) 三并环类衍生物抑制剂、其制备方法和应用
RU2781100C1 (ru) Соединения со спиро- и ароматическими кольцами и их применение
CN110229151B (zh) 吲嗪类化合物、其制备方法及用途
WO2023169170A1 (fr) Composé hétérocyclique utilisé en tant qu'inhibiteur de shp2, composition comprenant un composé hétérocyclique, et procédé l'utilisant
WO2022122037A1 (fr) Dérivé de dihydroisoquinolinone et son application

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200619

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

A4 Supplementary search report drawn up and despatched

Effective date: 20201201

RIC1 Information provided on ipc code assigned before grant

Ipc: C07D 401/14 20060101ALI20201125BHEP

Ipc: C07D 471/10 20060101ALI20201125BHEP

Ipc: A61P 35/00 20060101ALI20201125BHEP

Ipc: A61K 31/4427 20060101ALI20201125BHEP

Ipc: C07D 401/04 20060101ALI20201125BHEP

Ipc: C07D 487/04 20060101ALI20201125BHEP

Ipc: A61K 31/4985 20060101ALI20201125BHEP

Ipc: C07D 519/00 20060101ALI20201125BHEP

Ipc: A61P 35/02 20060101ALI20201125BHEP

Ipc: A61K 31/519 20060101ALI20201125BHEP

Ipc: A61P 35/04 20060101ALI20201125BHEP

Ipc: C07D 471/04 20060101AFI20201125BHEP

Ipc: A61K 31/497 20060101ALI20201125BHEP

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 35/04 20060101ALI20210714BHEP

Ipc: A61P 35/02 20060101ALI20210714BHEP

Ipc: A61P 35/00 20060101ALI20210714BHEP

Ipc: A61K 31/519 20060101ALI20210714BHEP

Ipc: A61K 31/4985 20060101ALI20210714BHEP

Ipc: A61K 31/497 20060101ALI20210714BHEP

Ipc: A61K 31/4427 20060101ALI20210714BHEP

Ipc: C07D 401/14 20060101ALI20210714BHEP

Ipc: C07D 401/04 20060101ALI20210714BHEP

Ipc: C07D 519/00 20060101ALI20210714BHEP

Ipc: C07D 487/04 20060101ALI20210714BHEP

Ipc: C07D 471/10 20060101ALI20210714BHEP

Ipc: C07D 471/04 20060101AFI20210714BHEP

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
INTG Intention to grant announced

Effective date: 20210920

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

Ref country code: AT

Ref legal event code: REF

Ref document number: 1472171

Country of ref document: AT

Kind code of ref document: T

Effective date: 20220315

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602019012229

Country of ref document: DE

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602019012229

Country of ref document: DE

Representative=s name: SIMMONS & SIMMONS LLP, DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2914852

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20220617

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220602

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220602

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1472171

Country of ref document: AT

Kind code of ref document: T

Effective date: 20220302

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220603

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220704

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220702

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602019012229

Country of ref document: DE

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

26N No opposition filed

Effective date: 20221205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20221130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221105

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221105

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20231026

Year of fee payment: 5

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231019

Year of fee payment: 5

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20231211

Year of fee payment: 5

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20231026

Year of fee payment: 5

Ref country code: IT

Payment date: 20231026

Year of fee payment: 5

Ref country code: FR

Payment date: 20231024

Year of fee payment: 5

Ref country code: DE

Payment date: 20231024

Year of fee payment: 5

Ref country code: CH

Payment date: 20231201

Year of fee payment: 5

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20220302